WO2005059118A2 - Stem cell targeting using magnetic particles - Google Patents

Stem cell targeting using magnetic particles Download PDF

Info

Publication number
WO2005059118A2
WO2005059118A2 PCT/GB2004/005156 GB2004005156W WO2005059118A2 WO 2005059118 A2 WO2005059118 A2 WO 2005059118A2 GB 2004005156 W GB2004005156 W GB 2004005156W WO 2005059118 A2 WO2005059118 A2 WO 2005059118A2
Authority
WO
WIPO (PCT)
Prior art keywords
use according
cell
particle
cells
magnetisable
Prior art date
Application number
PCT/GB2004/005156
Other languages
French (fr)
Other versions
WO2005059118A3 (en
Inventor
Jon Dobson
Alicia El Haj
Original Assignee
Keele University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Keele University filed Critical Keele University
Priority to AU2004299704A priority Critical patent/AU2004299704A1/en
Priority to US10/596,594 priority patent/US8469034B2/en
Priority to CA002550084A priority patent/CA2550084A1/en
Priority to EP04805976A priority patent/EP1699917A2/en
Priority to JP2006544539A priority patent/JP2007518710A/en
Publication of WO2005059118A2 publication Critical patent/WO2005059118A2/en
Publication of WO2005059118A3 publication Critical patent/WO2005059118A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/18Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes
    • A61K49/1896Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes not provided for elsewhere, e.g. cells, viruses, ghosts, red blood cells, virus capsides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0607Non-embryonic pluripotent stem cells, e.g. MASC
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6901Conjugates being cells, cell fragments, viruses, ghosts, red blood cells or viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P23/00Anaesthetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P23/00Anaesthetics
    • A61P23/02Local anaesthetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • A61P29/02Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID] without antiinflammatory effect
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82BNANOSTRUCTURES FORMED BY MANIPULATION OF INDIVIDUAL ATOMS, MOLECULES, OR LIMITED COLLECTIONS OF ATOMS OR MOLECULES AS DISCRETE UNITS; MANUFACTURE OR TREATMENT THEREOF
    • B82B3/00Manufacture or treatment of nanostructures by manipulation of individual atoms or molecules, or limited collections of atoms or molecules as discrete units
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N13/00Treatment of microorganisms or enzymes with electrical or wave energy, e.g. magnetism, sonic waves

Definitions

  • This invention relates to a novel method of magnetically manipulating stem cells ex vivo or in vivo and to methods of treatment related thereto.
  • stem cells in the form of a cell-based therapies is currently one of the most exciting and promising areas for disease treatment and reparative medicine.
  • basic research into the ways by which proliferation and differentiation of e.g. embryonic and adult stem cells can be controlled is vitally important.
  • US Patent No. 6,548,264 describes silica coated nanoparticles which comprise a magnetic metal core.
  • the magnetic core present in the particles enables the particles to be responsive to a magnetic field and therefore, the particles are suitable for use in diagnostic, imaging and recording systems.
  • the nanoparticles of the prior art may suffer from the disadvantage that they do not define the method of activation at a cellular level.
  • Magnetic bead twisting cytometry has been used to define the mechanical properties of single cells and to demonstrate that external mechanical forces can be transmitted across the cell surface and through the cytoskeleton via transmembrane cell adhesion molecules such as integrins, see, for example, Wang, N and Ingberger, DE (1995) Probing transmembrane mechanical coupling and cytomechanics using magnetic twisting cytometry. Biochem. Cell Biol. 73: 327-335.
  • Bone marrow contains multipotential stromal stem cells or mesenchymal stem cells which can differentiate into, ter alia, fibroblastic, osteogenic, adipogenic and reticular cells.
  • mesenchymal stem cells such as human bone marrow stromal fibroblasts can be isolated from volunteer donors and may retain their multilineage (adipocytic, chondrogenic, osteoblastic) potential.
  • One advantage in the use and manipulation of the aforementioned cells lies in their lack of immunogenicity which provides the potential for use of these cells in, inter alia, cartilage and bone repair.
  • Figure 2 shows the results of experiments using bone marrow stromal cells with internal calcium levels up- regulated as a result of the application of magnetic fields to magnetic nanoparticles attached to a His-tagged TREK channel.
  • stem cells tagged with magnetic nanoparticles can be delivered to or held at, a particular repair site by external magnetic manipulation.
  • these concepts further to include remote activation of specific cellular membrane receptors, which in essence, involves localising cells e.g. stem cells. More simply this involves deposition of stem cells at a site e.g. a repair site, retaining the cells at the site and remotely activating the cells in situ vvithin the patient.
  • the present invention addresses issues of targeting specific receptors on cells for remote activation of transmembrane ion channels in stem cells.
  • magnetic nanoparticle-based technologies are increasingly used clinically, in many facets of healthcare e.g. contrast enhancement for MRI.
  • the achieved differentiation acts as a model for binding strategies which allows both remote targeting within the body and/or activation at specific sites when localised.
  • the present invention enables the targeting of a variety of stem cell receptor types, such as mechano-activated ion channels e.g. K+ channels (TREK), calcium channels, integrins and surface membrane binding sites such as RGD, present in human bone marrow stem cells.
  • stem cell receptor types such as mechano-activated ion channels e.g. K+ channels (TREK), calcium channels, integrins and surface membrane binding sites such as RGD, present in human bone marrow stem cells.
  • TREK mechano-activated ion channels
  • calcium channels e.g. K+ channels (TREK)
  • integrins e.g., calcium channels
  • RGD surface membrane binding sites
  • the targeting of other known receptors such as external growth factors (e.g. TGFB and BMP2) which have been shown to activate downstream transcription factors such as Runx2 and Osterix, critical for stem cell differentiation can also be achieved.
  • TGFB and BMP2 external growth factors
  • the present invention provides the opportunity for true engraftment of, inter alia, human mesenchymal stem cells, long-term biological effects on the stem cells at the site of injury or repair. Furthermore, the ability to select, expand and differentiate these cells and target the cells using magnetic nanoparticles is especially advantageous. Furthermore, utilisation of the present invention provides therapeutic implications in, inter alia, gene therapy and tissue engineering.
  • Biocompatible magnetic nanoparticles primarily composed of a magnetite (Fe 3 O ) and/or maghemite (Fe O 3 ) core with either a silica, dextran, or PNA coating may be utilised in the present invention.
  • Such particles may be synthesized following methods known in the art. However, it will be understood that other magnetic nanoparticles may be utilised. Particle sizes can range from -lOnm up to a few microns e.g. 1 to lO ⁇ m. Commercially available magnetic micro- and nanoparticles with varying surface chemistry may also be used.
  • the coatings may be functionalized and crosslinked to membrane attachment motifs such as those described above.
  • the magnetic nanoparticles may be modified so as to customise, inter alia, particle internalization frequency and binding efficiency and stability will be examined as will the effects of binding on cell viability and function. Modification may also include customisation of internal binding sites as well as sites on the outer membrane.
  • a variety of coatings may be used in magnetic nanoparticle binding and loading in human osteoblasts 14 ' 15 and these techniques may be further optimized for stem cell binding, delivery and activation e.g. using adult primary marrow human stem cells and/or human embryonic stem cells.
  • high gradient magnets e.g. external rare earth (primarily NdFeB), high-gradient magnets
  • NdFeB external rare earth
  • high-gradient magnets may be used to target the stem cells to specific sites within an in vitro test system and/or in vivo.
  • Such magnets produce high field/gradient products which exert a translational force on the magnetic particles loaded onto the cells, holding them at the target site according to the equation:
  • Remote mechanical activation may be achieved using e.g. a magnetic conditioning bioreactor.
  • bioreactors which are known per se, enable forces to be applied to magnetic particles attached to cells cultured in vitro within a multi-well 2D system or in vivo a 3D scaffold-based system.
  • Stem cells e.g. Mesenchymal stem cells and populations generated therefrom, such as osteogenic, chondrogenic and adipogenic populations may be isolated using, for example, magnetic activated cell sorting (MACS) with a monoclonal antibody e.g. STRO-1 using standard protocols 'known per se x ⁇
  • MCS magnetic activated cell sorting
  • STRO-1 monoclonal antibody
  • Such protocols include those known for BMSc culture in monolayer .and using 3D scaffolds composed of biodegradable polymers such as poly lactic acid 91
  • PLLA collagen gels
  • a remote manner it is intended to mean, e.g. a non-contacting manner and in the case of in vivo activating/targeting specifically from outside the body.
  • a method of magnetically manipulating a stem cell in vivo or in vitro which comprises the association of a magnetisable particle with a stem cell.
  • the method may comprise ex vivo manipulation of an in vivo process.
  • a reference to a cell shall be construed to include a plurality of cells.
  • the invention provides a method as hereinbefore described which comprises the activation and/or targeting of a magnetisable particle with a stem cell as hereinbefore described.
  • a method of magnetically manipulating a stem cell which comprises the association of a magnetisable particle with a cell characterised in that the method comprises agonising or antagonising ion channels within a cell by the association of a magnetisable particle with a cell.
  • the magnetisable particle may be associated directly with the cell.
  • the method may comprise associating the magnetisable particle with an antibody, enzyme, etc., which is subsequently associated with the cell.
  • the association of a magnetisable particle with a cell may comprise the introduction of such a particle into a cell, the attachment of such a particle to a cell, e.g. externally or internally to a cell, or any combination thereof.
  • the magnetisable particles may be associated intracellularly or extracellularly or a combination of intracellularly and extracellularly.
  • the particles are associated intracellularly.
  • this will comprise association with an internal binding site.
  • the particle(s) may be associated with the N-te ⁇ riinus region of the ion channel.
  • the particle(s) may be associated with the COOH terminus region of the ion channel.
  • ion channels and binding sites may be utilised in the method of the invention.
  • internal binding sites which correspond to the N-terminus region of the ion channel, as seen in TREK- lor which corresponds to the COOH terminus region ofthe ion channel, as seen in TREK-1 may be utilised as well as other binding sites known per se.
  • the method of the invention may comprise the manipulation of mammalian cells or other cell types, such as bacterial cells, plant cells, etc. However, it will be understood by the skilled man that the method of the present invention may be used to manipulate other cell types not mentioned herein. Furthermore, the method may be an in vitro method or an in vivo method, although an in vivo method is preferred.
  • the method of the invention may comprise the up-regulation or down-regulation of gene expression in stem cells in response to mechanical manipulation of the stem cells as described herein.
  • the stem cells may be induced to follow particular differentiation pathways such as described herein.
  • the method of the invention comprises the remote manipulation of cells and/or of agonising or autagonising ion channels, e.g. manipulation from outside the body., i.e. remote mechanical activation.
  • the method ofthe invention may be utilised in relation to a variety of cells which are known per se. However, preferentially, the method is suitable for use with mammalian stem cells.
  • the method of the invention may be utilised in connection with any conventionally known ion channels within the cell which are hereinbefore described.
  • the method is especially suited for use in mechanosensitive ion channels.
  • mechanosensitive ion channels have been identified in many cell types and have been predominantly described as calcium or potassium ion channels, although it should be understood that the method of the invention is not limited to use in relation to calcium or potassium ion channels.
  • one such channel which has been well characterised at the molecular level and at the functional level in neuronal cells is the chromosomal gene TREK-1, which is part of the 2P K+ channel family.
  • TREK-1 channels have been identified in bone cells, and are known to respond to shear stress, cell swelling and membrane stretch as well as other external agents such as fatty acids and general anaesthetics.
  • a particular aspect of the present invention is to provide a method of manipulating mechanosensitive ion channels.
  • these channels are instrumental in normal cellular function and play a particularly important role in, for example, the production of bone and connective tissue or activation ofthe peripheral nervous system, the ability to manipulate them remotely, e.g. from outside the body, is especially advantageous an provides applications in, ter alia, pain relief, e.g. anaesthetics, therapeutics, tissue engineering and repair and cancer therapy.
  • the method may also be suitable for use with conventionally non mechanosensitive cells and/or ion channels by the transfection of channels into cells which may otherwise be otherwise non-responsive.
  • Noltage-gated and ligand-gated ion channels are also "mechanoresponsive" in that they respond to mechanical stresses on the ion channel generated by coulomb forces (in the case of voltage-gated channels) and binding forces (in the case of ligand-gated channels). As such, all ion channels can be activated by the method described herein provided that the magnetisable particle is coupled, either directly or indirectly, to the mechanoresponsive region ofthe channel protein.
  • the ion channel is a voltage-gated ion channel, alternatively, the ion channel is a ligand-gated ion channel.
  • the magnetisable particle used in the method ofthe invention may be inherently magnetic or, alternatively, may be one which reacts in a magnetic field.
  • any magnetic material may be used, however, by the term magnetic we mean, for example, a material which is paramagnetic superparamagnetic, ferromagnetic and/or antiferromagnetic, examples of which include elemental iron (Fe), or an compound, e.g. an iron salt, such as, magnetite (Fe 3 O 4 ), maghemite ( ⁇ Fe 2 O 3 ), and greigite (Fe 3 S 4 ), or a chromium compound, e.g.
  • the magnetic material comprises particles, e.g. nanoparticles, which comprises a magnetic core with a biocompatible coating.
  • particles e.g. nanoparticles, which comprises a magnetic core with a biocompatible coating.
  • such preferred particles are nanoparticles and especially nanoparticles having a core and, e.g. a silica shell enveloping the core.
  • porous particles with multiple magnetic centres within the pores are those nanoparticles described in US Patent No. 6,548,264 which is incorporated herein by reference.
  • the prior art nanoparticles may have a mean size of less than 1 micron, each of said nanoparticles comprising (a) a core comprising a magnetisable particle and (b) a silica shell enveloping the core, wherein the magnetisable particle is a magnetic material as hereinbefore described.
  • the micro- and nano- particles (intended to be attached to the cells) will generally be substantially spherical or elliptical.
  • the size of the particles may vary according, inter alia, to the nature of the magnetisable material, the application, etc.
  • an example of particles may be nanoparticles can having a mean size, e.g. diameter, of 5000 nm or less, e.g. from 1 nm to 5000 nm, preferably from 1 nm to 1000 nm, more preferably from 1 nm to 300 nm, or from 2 nm to 10 nm).
  • the particles for attachment to the cells may be coated or uncoated and single or multi-domain.
  • suitable particles include, but are not limited to:
  • the ion channels may be activated by attaching the magnetisable particles as hereinbefore described to specific regions of the cellular membrane and/or to specific "receptors" on the ion channels themselves.
  • the mechanical forces required to activate the channels can then be applied remotely by a magnetic field acting on these magnetic particles.
  • the method of the invention comprises modifying a magnetisable particle as hereinbefore described by tagging the particle with one or more specific antibodies or protein binding motifs which recognise key cellular elements within a cell.
  • transmembrane adhesion molecules such as integrins, cadherins, selectins, and immunoglobulins or dispersed membrane adhesion proteins such as RGD (argiiiine-glycine-aspartate), see, for example, . J. Chen, B. Fabry, E. L. Schiffrin, and N. Wang (2001) Twisting integrin receptors increases endothelin-1 gene expression in endothelial cells Am JPhysiol Cell Physiol. 280: 1475-84 ; A. R. Bausch, U. Hellerer, M. Essler, M. Aepfelbacher, and E.
  • the method ofthe invention is especially advantageous because it provides a method of treatment of a variety of disorders. Indeed the invention provides a method of treatment which is applicable to any disorder in which one or more ion channels play a role. In addition, the invention provides a method for potential control of ion channel activation including pain relief, e.g. an anaesthetic role.
  • the method of treatment as hereinbefore described should not be considered to be limited, but it is especially advantageous in tissue and/or bone repair.
  • the method of treatment can be to facilitate further treatment by providing a method of pain relief, e.g. for localised anaesthesia, to targeted regions ofthe body.
  • the nature of such cells may vary depending upon the nature ofthe tissue of interest.
  • the cells may be ligamentum cells for growing new ligaments, tenocytes for growing new tendon.
  • the cells may be chondrocytes and/or other stromal cells, such as chondrocyte progenitor cells.
  • the method of the invention may include the regeneration of tissue or the generation of artificial tissue, such as skin, cartilage, ligament, tendon, muscle or bone.
  • the method may comprise wound healing and/or tissue adhesion.
  • the method may comprise bone repair and/or bone growth.
  • the method of the invention may include, for example, dental applications and/or veterinary applications.
  • the method also may be used as a mechanism for selectively killing cells (such as tumour cells) in vivo.
  • magnetisable particles are attached to the target cell membrane or ion channel protein and a magnetic field is applied to the in vivo target region.
  • the rapid, cyclic opening and closing (via the application of a time varying magnetic field), and/or the holding open (via the application of a static magnetic field) of ion channels in the cell membrane allows ions (such as Ca " " " ) to flood the cell, inducing osmotic shock and, consequently, cell death.
  • a method of destroying cells or inhibiting cell growth which comprises agonising or antagonising ion channels within a cell which by the association of a magnetisable particle with a cell.
  • the method may comprise a method of inducing osmotic shock to a cell, e.g. by agonising or antagonising ion channels within a cell by the association of a magnetisable particle with a cell.
  • the method is especially useful in the treatment or alleviation of a tumour cell, e.g. a cancer cell.
  • the method may comprise the killing of cells by holding ion channels open with a targeted static magnetic field.
  • the method may comprise the killing of cells via cyclically opening and closing ion channels with a targeted, time- varying magnetic field.
  • the magnetic field may be varied depending upon, inter alia, the nature of the disorder to be treated, but may be, for example, at a frequency of from 0.1 to 10 Hz. But, frequencies outside this range can also be used.
  • the magnetic field will typically have a flux density in the order of (but not limited to) 10 mT to 1400 mT.
  • the magnetic field may be generated outside the body for the case of in vivo applications, and may be provided by a permanent magnet or an electromagnet.
  • the magnetic field may be a constant or a variable field, e.g. a permanent magnet may be moved relative to the cells.
  • a magnetic field may be generated by provision of appropriate electric current levels to the electromagnetic, optionally, in combination with alternating current.
  • a method of inducing a therapeutic effect in a cell which comprises agonising or antagonising ion channels within the cell by the association of a magnetisable particle with the cell and magnetically manipulating the magnetisable particle.
  • a method of treatment which comprises the administration of a therapeutically active agent which may be administered simultaneously, separately or sequentially with a magnetisable particle whilst agonising or antagonising ion channels vvithin the cell.
  • the use may comprise ex vivo manipulation of an in vivo process. More particularly, the invention provides the use of a magnetisable particle in the manufacture of a system for magnetically manipulating a cell which system comprises the association of a magnetisable particle with a cell and agonising or antagonising ion channels witliin the cell.
  • the magnetisable particle may be associated directly with the cell.
  • the use may comprise associating the magnetisable particle with an antibody, enzyme, etc., which is subsequently associated with the cell.
  • the particle(s) may be associated with the N-terminus region of the ion channel.
  • the particle(s) may be associated with the COOH terminus region ofthe ion channel.
  • the use ofthe invention may comprise the manipulation of mammalian cells or other cell types, such as bacterial cells, plant cells, etc.
  • the use may be an in vitro use or an in vivo use, although an in vivo use is preferred.
  • the use of the invention comprises the remote manipulation of cells and/or of agonising or autagonising ion channels, e.g. manipulation from outside the body, i.e. remote mechanical activation.
  • the use of the invention may be utilised in relation to a variety of cells, which are known per se.
  • the use is suitable for use with mammalian somatic cells, for example, bone, cartilage, muscle (skeletal and cardiac) lymphatic cells, endocrine cells, urinary system cells, cells relating to the reproduction system, neuronal cells and tumour cells.
  • the use of the invention may be utilised in connection with any conventionally known ion channels within the cell, which is hereinbefore described.
  • the use is especially suited for use in mechanosensitive ion channels hereinbefore described.
  • a particular aspect of the present invention is to provide the use in the manufacture of a system for manipulating mechanosensitive ion channels.
  • the use may also be suitable for use with conventionally non mechanosensitive cells and/or ion channels by the transfection of channels into cells which may otherwise be otherwise non-responsive.
  • the ion channel is a voltage-gated ion channel, alternatively, the ion channel is a ligand-gated ion channel.
  • any magnetisable material may be used, examples of which include elemental iron (Fe), or an iron compound, e.g. an iron salt, such as, magnetite (Fe 3 O 4 ), maghemite ( ⁇ Fe 2 O 3 ), and greigite (Fe 3 S 4 ), or a chromium compound, e.g. a chromium salt, such as, chromium oxide (CrO 2 ), or any combination thereof.
  • the magnetic material comprises particles which comprises a magnetic core with a biocompatible coating.
  • such preferred particles are nanoparticles and especially nanoparticles having a core and, e.g. a silica shell enveloping the core.
  • porous particles with multiple magnetic centres within the pores are those nanoparticles described in US Patent No. 6,548,264 which is incorporated herein by reference.
  • the use of the invention comprises modifying a magnetisable particle as hereinbefore described by tagging the particle with one or more specific antibodies or protein binding motifs which recognise key cellular elements within a cell.
  • These include transmembrane adhesion molecules, such as integrins, cadherins, selectins, and immunoglobulins or dispersed membrane adhesion proteins such as RGD (arginine-glycine-aspartate) .
  • the use of the invention is especially advantageous because it provides a system suitable for use in the treatment of a variety of disorders.
  • the invention provides the use in the manufacture of a medicament suitable for a treatment, which is applicable to any disorder in which one or more ion channels play a role.
  • the invention provides the use for potential control of ion channel activation including pain relief, e.g. an anaesthetic role.
  • a magnetisable particle in the manufacture of a medicament suitable for the treatment of a patient suffering from a disorder in which an ion channel plays a role which comprises the administration to such a patient of magnetisable particles as hereinbefore described and manipulating those particles using a magnetic field.
  • the use as hereinbefore described should not be considered to be limited, but it is especially advantageous in tissue and/or bone repair.
  • the use can be to facilitate further treatment by providing a method of pain relief, e.g. for localised anaesthesia, to targeted regions ofthe body.
  • the nature of such cells may vary depending upon the nature ofthe tissue of interest.
  • the cells may be ligamentum cells for growing new ligaments, tenocytes for growing new tendon.
  • the cells may be chondrocytes and/or other stromal cells, such as chondrocyte progenitor cells.
  • the use may include the regeneration of tissue or the generation of artificial tissue, such as skin, cartilage, ligament, tendon, muscle or bone.
  • the use may comprise wound healing and/or tissue adhesion.
  • the use may comprise bone repair and/or bone growth.
  • the use ofthe invention may include, for example, dental applications and/or veterinary applications.
  • the use also may be used as a mechanism for selectively killing cells (such as tumour cells) in vivo as hereinbefore described.
  • a magnetisable particle in the manufacture of a system for destroying cells or inhibiting cell growth which comprises agonising or antagonising ion channels within a cell which by the association of a magnetisable particle with a cell.
  • the use may comprise use in a method of inducing osmotic shock to a cell, e.g. by agonising or antagonising ion channels within a cell by the association of a magnetisable particle with a cell.
  • the use in this aspect of the invention is especially useful in the treatment or alleviation of a tumour cell, e.g. a cancer cell.
  • the use may comprise the killing of cells by holding ion channels open with a targeted static magnetic field.
  • the use may comprise the killing of cells via cyclically opening and closing ion channels with a targeted, time- varying magnetic field.
  • a magnetisable particle in the manufacture of a system for inducing a therapeutic effect in a cell which comprises agonising or antagonising ion channels within the cell by the association of a magnetisable particle with the cell and magnetically manipulating the magnetisable particle.
  • a magnetisable particle in the manufacture of a system comprising a therapeutically active agent which may be administered simultaneously, separately or sequentially with the magnetisable particle whilst agonising or antagonising ion channels within the cell.
  • a magnetisable particle in the manufacture of a system for targeting a therapeutically active agent to a cell which comprises agonising or antagonising ion channels within the cell by the association of a magnetisable particle with the cell, magnetically manipulating the magnetisable particle and simultaneously, separately or sequentially administering the therapeutically active agent.
  • kits comprising a therapeutically active agent and means for associating a magnetisable particle with a cell.
  • the kit may comprise a vessel containing a therapeutically active agent, a source of magnetisable particles and instructions for the simultaneous, sequential or separate a ⁇ 'ministration thereof.
  • the kit of the invention may also include other agents known er se.
  • the invention may also include the use of a kit as hereinbefore described in the manufacture of a medicament.
  • Figure lb illustrates primary human astrocytes with membrane bound RGD coated carboxyl ferromagnetic particles (4 ⁇ m) (magnification x 1000);
  • Figure 2 is a schematic of the TREK ion channel showing structure and location of the His. tags present in the protein. Red circles indicate the sites ofthe His tags at the three sites, the primary loop, the COOH terminus and the NH terminus;
  • Figure 3 is a representation of the magnetic activation of Trek-1 monitored via downstream changes in intracellular calcium
  • Figure 4 is a representation of the magnetic activation of TREK-1 induces transient rise in intracellular calcium in HEK293 T cells co-transfected with and Flashpericam.
  • the model system consists of a peristaltic pump connected to tubing which feeds into channels witnin agar gel blocks.
  • the magnets can be placed at various positions in relation to the channels and the magnetic field and gradient at the target site is measured using an axial Hall probe interfaced to a gaussmeter.
  • the magnetic fields generated by the rare earth magnets will be characterised using a Redcliffe Diagnostics MagScan field mapping system requested for this project.
  • the gel channel will be excised and assayed for cell capture using staining techniques. Magnetic particle capture will be quantified by performing Superconducting Quantum Interference Device (SQUID) magnetometry measurements on the freeze dried gel blocks.
  • Models may be used to optimize the delivery and targeting parameters, such as magnetic field strength and geometry, magnetic particle characteristics, number of particles per cell, etc.
  • scaffolds are seeded with 10 6 -10 9 BMSc dependant on scaffold size and cultured for 24 hours prior to placing within the bioreactor. Constructs are then subjected to varying magnetic loading regimes, e.g. 1 hour at 1 Hz frequency with forces ranging from 1-lOOpN per particle. These parameters are controllable and will allow optimisation of the system for varying cell types and scaffold materials. Following treatment, cells may be removed and subjected to RNA and protein analysis at varying points after activation.
  • osteoblastic transcription factors such as runx 2 and osterix
  • matrix proteins such as osteopontin, collagen type 1, alkaline phosphatase and osteocalcin.
  • Animal trials of this technology support the ability to remotely activate stem cells to promote bone call differentiation and new bone formation by cells held in vivo within subcutaneous diffusion chambers using a mouse SCID model. In this way, comparisons can be made with in vitro experiments. Targeting of cells to specific tissues in vivo may also be advised.
  • Human-derived osteoprogenitors from mesenchymal stem cells may be used.
  • In vivo bone formation may be assessed using the subcutaneous implant model in severely compromised immunodeficient (SCID) mice and the diffusion chamber model.
  • SCID immunodeficient
  • This provides a rapid and robust model to validate, in vivo, the efficacy for targeting of magnetic micro- and nanoparticles and provides a clear demonstration of bone formation.
  • the diffusion chamber assay provides unequivocal demonstration of bone formation by implanted cells as opposed to host cells.
  • the subcutaneous implant model remains the industry standard for the assessment of skeletal tissue formation and one of us (RO) has published on the use of both the sc and DC models under a project license to RO (30/1759) for assessment of skeletal tissue engineering 22 .
  • selected human osteoprogenitor cells will be implanted subcutaneously in SCID mice for four weeks while for diffusion chamber studies, cells and magnetic particle composites will be placed into each diffusion chamber and the chambers implanted intraperitoneally into athymic nude mice (MFI-nu-nu; 4-6 weeks old; Harlan UK Ltd) for 10 weeks. Thereafter, diffusion chambers will be removed, fixed overnight (95% ethanol, 4°C) and embedded undecalcified in poly(hydroxymethylmethacrylate) resin at 4°C. New bone formation will be assessed by histological techniques including frozen, paraffin and methylmethacrylate plastic sections.
  • Magnetic microparticles (d-4 ⁇ m) were coated with a biotinylated ⁇ 2 / ⁇ -l subunit of a voltage gated calcium ion channel receptor antibody. After 4 days, particles were attached to the stem cells for 40 minutes via the calcium channel receptor. After 40 minutes, the cells were exposed to a 1Hz magnetic field which applied a force of approximately 30 picoNewtons per particle ( ⁇ 2 particles/cell). After 2 hours 40 minutes, the particles were detached from the cells and removed by aspiration. The original culture media was returned to the samples which were then further cultured for another 24 hour period. RNA from the control and stimulated groups was collected at day five. Gene microarray analysis was performed on each of the samples. 8000 genes/sample were analysed using HG-Focus human genome chips (Affymetrix UK Ltd) in response to magnetic activation (upregulation and downregulation taken as two fold increase/decrease).
  • Microarray data from these experiments showed that the mechanical stimulation resulted in the downregulation of certain genes such as nerve growth factor and fibroblast growth factor (Table 1). This is an indication that the application of mechanical force using magnetic particles is guiding the stem cell differentiation away from the neuronal and fibroblast pathways.
  • the upregulation of genes such as tetranectin in response to the mechanical force application indicates a differentiation of the cells towards an osteogenic pathway. Upregulation of genes involved in cytoskeletal reorganisation and cell adhesion proteins correlate with expected cell processes after force application.

Abstract

The present invention provides a method of selectively activating and/or targeting stem cells which enables the cells to then be manipulated mechanically in a remote manner wherein the method comprises magnetically manipulating a stem cell in vivo or in vitro by the association of a magnetisable particle within the stem cell.

Description

Method
This invention relates to a novel method of magnetically manipulating stem cells ex vivo or in vivo and to methods of treatment related thereto.
The use of stem cells in the form of a cell-based therapies is currently one of the most exciting and promising areas for disease treatment and reparative medicine. Clearly, basic research into the ways by which proliferation and differentiation of e.g. embryonic and adult stem cells can be controlled is vitally important.
US Patent No. 6,548,264 describes silica coated nanoparticles which comprise a magnetic metal core. The magnetic core present in the particles enables the particles to be responsive to a magnetic field and therefore, the particles are suitable for use in diagnostic, imaging and recording systems. However, the nanoparticles of the prior art may suffer from the disadvantage that they do not define the method of activation at a cellular level.
Magnetic bead twisting cytometry has been used to define the mechanical properties of single cells and to demonstrate that external mechanical forces can be transmitted across the cell surface and through the cytoskeleton via transmembrane cell adhesion molecules such as integrins, see, for example, Wang, N and Ingberger, DE (1995) Probing transmembrane mechanical coupling and cytomechanics using magnetic twisting cytometry. Biochem. Cell Biol. 73: 327-335.
There have been many developments in biocompatible magnet nanoparticle synthesis, characterization1"3 and applications of novel magnetic techniques in the field of healthcare4"6. This work primarily has involved investigating the controlled and directed transport of pharmaceuticals. In these systems therapeutic drugs or genes may be attached to magnetic carrier particles (usually polymer coated magnetite), which are then concentrated at the target site in vivo by the application of spatially focused, high gradient magnetic fields. Once the drug/carrier complexes have accumulated at the target site, the drug is released and uptake at the sites is enhanced. Investigations have been made into new methods for magnetic targeting for gene therapy as well as theoretically and experimentally examining and improving deposition of magnetic micro- and nanoparticle carriers in model systems in vitro and in vivo ' .
Short-term experiments where force is applied to the cell membrane using torque or where tension is applied to transmembrane proteins such as RGD or collagen molecules has been described by a number of researchers ' . These experiments use 'mechanical' stimulation of the membrane to trigger short term internal calcium fluxes in a variety of cells. It is known that mechanical signalling using other techniques can trigger differentiation pathways in bone marrow stromal cells down the osteogenic lineage and in particular, that low level mechanical signals across the membrane can up-regulate expression and DNA binding activity of osteoblastic specific transcription factors, cbfal and cfos12'13.
In these investigations, force can be applied to a number of different tagged receptors. It has been demonstrated how we can influence downstream processes and enhance collagen and other matrix protein synthesis15. Using bone marrow derived mesenchymal stem cells conditioned to differentiate along the osteogenic and chondrogenic lineage we have been . investigating downstream gene regulation in response to magnetic particle activation of specific receptors. Preliminary data has shown an up-regulation in Runx 2 in response to magnetic particle stimulation of calcium channels in human mesenchymal stem cells followed by up-regulation of a mechanosensitive matrix protein, osteopontin. In addition, we have evidence of up- regulation of SOX 9 following stimulation of monolayer human dedifferentiated chondrocytes. These studies have been extended to 3D analysis of cell-seeded scaffolds over long-term culture to investigate the use of these strategies for construct fabrication in tissue engineering in vitro. Furthermore, preliminary studies which include a dose-response analysis of particle number and force applied are encouraging and indicate increased matrix synthesis and expression ofthe osteogenic phenotype14.
Bone marrow contains multipotential stromal stem cells or mesenchymal stem cells which can differentiate into, ter alia, fibroblastic, osteogenic, adipogenic and reticular cells. These mesenchymal stem cells, such as human bone marrow stromal fibroblasts can be isolated from volunteer donors and may retain their multilineage (adipocytic, chondrogenic, osteoblastic) potential. One advantage in the use and manipulation of the aforementioned cells lies in their lack of immunogenicity which provides the potential for use of these cells in, inter alia, cartilage and bone repair.
Our as yet unpublished co-pending International Patent Application, No. PCT/GB2003/ 002624 combines the magnetic nanoparticle approach with knowledge of mechanosensitive ion channels, in particular, the TREK K+ channel. It is established that the TREK channel is present in osteogenic, chondrogenic and bone marrow stromal cells In order to define more closely the targeting of specific receptors to control activation, we have used HIS-tagged clones of the TREK gene. HIS tags have been inserted into particular regions of the TREK molecule to allow attachment of HIS antibody or NI2+ bound magnetic particles which can then be remotely torques using a magnetic field. Sites of the ion channel protein which lie both internal and external to the cell membrane have been tagged and in this way we can identify the mechanosensitive regions ofthe molecule as well as define the signal frequencies required to switch on downstream processes. Figure 2 shows the results of experiments using bone marrow stromal cells with internal calcium levels up- regulated as a result of the application of magnetic fields to magnetic nanoparticles attached to a His-tagged TREK channel.
It has been shown that conditioning connective tissue cells in vitro can be achieved, by, inter alia, the development of a magnetic force bioreactor which enables magnetic fields to be applied in vitro to 2D monolayer cultures and 3D cell-seeded scaffolds. However, neither US '264 nor Wang solve or even address the problems surrounding two fundamental questions which need to be addressed, and which encompass the ultimate goal of engineering cells for clinical use, namely;
(i) how will cells be targeted to the site of repair and held at that site; and
(ii) how will cells e.g. stem cells, be conditioned or differentiated in vitro and/or in vivo.
We have now surprisingly found ways by which stem cells tagged with magnetic nanoparticles can be delivered to or held at, a particular repair site by external magnetic manipulation. In addition, we have developed these concepts further to include remote activation of specific cellular membrane receptors, which in essence, involves localising cells e.g. stem cells. More simply this involves deposition of stem cells at a site e.g. a repair site, retaining the cells at the site and remotely activating the cells in situ vvithin the patient.
In particular, the present invention addresses issues of targeting specific receptors on cells for remote activation of transmembrane ion channels in stem cells. Importantly, magnetic nanoparticle-based technologies are increasingly used clinically, in many facets of healthcare e.g. contrast enhancement for MRI.
In the present invention we have achieved early stages of differentiation of these cell types. Moreover, the achieved differentiation acts as a model for binding strategies which allows both remote targeting within the body and/or activation at specific sites when localised.
Thus, the present invention enables the targeting of a variety of stem cell receptor types, such as mechano-activated ion channels e.g. K+ channels (TREK), calcium channels, integrins and surface membrane binding sites such as RGD, present in human bone marrow stem cells. Importantly, such receptors have the potential for remote activation. The targeting of other known receptors, such as external growth factors (e.g. TGFB and BMP2) which have been shown to activate downstream transcription factors such as Runx2 and Osterix, critical for stem cell differentiation can also be achieved.
Thus, the present invention provides the opportunity for true engraftment of, inter alia, human mesenchymal stem cells, long-term biological effects on the stem cells at the site of injury or repair. Furthermore, the ability to select, expand and differentiate these cells and target the cells using magnetic nanoparticles is especially advantageous. Furthermore, utilisation of the present invention provides therapeutic implications in, inter alia, gene therapy and tissue engineering.
Biocompatible magnetic nanoparticles, primarily composed of a magnetite (Fe3O ) and/or maghemite (Fe O3) core with either a silica, dextran, or PNA coating may be utilised in the present invention. Such particles may be synthesized following methods known in the art. However, it will be understood that other magnetic nanoparticles may be utilised. Particle sizes can range from -lOnm up to a few microns e.g. 1 to lOμm. Commercially available magnetic micro- and nanoparticles with varying surface chemistry may also be used. The coatings may be functionalized and crosslinked to membrane attachment motifs such as those described above. The magnetic nanoparticles may be modified so as to customise, inter alia, particle internalization frequency and binding efficiency and stability will be examined as will the effects of binding on cell viability and function. Modification may also include customisation of internal binding sites as well as sites on the outer membrane. A variety of coatings may be used in magnetic nanoparticle binding and loading in human osteoblasts14'15 and these techniques may be further optimized for stem cell binding, delivery and activation e.g. using adult primary marrow human stem cells and/or human embryonic stem cells. Targeting
Conventionally known high gradient magnets, e.g. external rare earth (primarily NdFeB), high-gradient magnets, may be used to target the stem cells to specific sites within an in vitro test system and/or in vivo. Clearly, it is a preferred aspect of the invention to target the stem cells in vivo. Such magnets produce high field/gradient products which exert a translational force on the magnetic particles loaded onto the cells, holding them at the target site according to the equation:
Fmag = (X2~X1)V- B( VB)
Activation
Remote mechanical activation may be achieved using e.g. a magnetic conditioning bioreactor. Such bioreactors, which are known per se, enable forces to be applied to magnetic particles attached to cells cultured in vitro within a multi-well 2D system or in vivo a 3D scaffold-based system. Stem cells, e.g. Mesenchymal stem cells and populations generated therefrom, such as osteogenic, chondrogenic and adipogenic populations may be isolated using, for example, magnetic activated cell sorting (MACS) with a monoclonal antibody e.g. STRO-1 using standard protocols 'known per sex\ Such protocols include those known for BMSc culture in monolayer .and using 3D scaffolds composed of biodegradable polymers such as poly lactic acid 91
(PLLA) or collagen gels .
We have now found a method of selectively activating and/or targeting stem cells which enables the cells to then be manipulated mechanically in a remote manner.
By the term "in a remote manner" it is intended to mean, e.g. a non-contacting manner and in the case of in vivo activating/targeting specifically from outside the body. Thus according to the invention we provide a method of magnetically manipulating a stem cell in vivo or in vitro which comprises the association of a magnetisable particle with a stem cell.
The method may comprise ex vivo manipulation of an in vivo process. Furthermore, it will be understood by the skilled man that a reference to a cell shall be construed to include a plurality of cells.
More particularly, the invention provides a method as hereinbefore described which comprises the activation and/or targeting of a magnetisable particle with a stem cell as hereinbefore described.
According to a further aspect of the invention we provide a method of magnetically manipulating a stem cell which comprises the association of a magnetisable particle with a cell characterised in that the method comprises agonising or antagonising ion channels within a cell by the association of a magnetisable particle with a cell.
According to a yet further aspect of the invention we provide a method as hereinbefore described which include a differentiation step.
In this aspect of the invention the magnetisable particle may be associated directly with the cell. Alternatively, the method may comprise associating the magnetisable particle with an antibody, enzyme, etc., which is subsequently associated with the cell.
The association of a magnetisable particle with a cell may comprise the introduction of such a particle into a cell, the attachment of such a particle to a cell, e.g. externally or internally to a cell, or any combination thereof. Thus, the magnetisable particles may be associated intracellularly or extracellularly or a combination of intracellularly and extracellularly. However, in a preferred aspect of the invention the particles are associated intracellularly. When the method of the invention comprises intracellular association this will comprise association with an internal binding site. By way of example only, for TREK-1, the particle(s) may be associated with the N-teπriinus region of the ion channel. Alternatively, the particle(s) may be associated with the COOH terminus region of the ion channel. It will be appreciated by one skilled in the art that numerous ion channels and binding sites may be utilised in the method of the invention. Thus, internal binding sites which correspond to the N-terminus region of the ion channel, as seen in TREK- lor which corresponds to the COOH terminus region ofthe ion channel, as seen in TREK-1 may be utilised as well as other binding sites known per se.
Thus, we also provide a method of manipulating a mechanosensitive ion channel characterised in that the method comprises the association of a magnetisable particle with an ion channel, either directly or indirectly.
The method of the invention may comprise the manipulation of mammalian cells or other cell types, such as bacterial cells, plant cells, etc. However, it will be understood by the skilled man that the method of the present invention may be used to manipulate other cell types not mentioned herein. Furthermore, the method may be an in vitro method or an in vivo method, although an in vivo method is preferred.
The method of the invention may comprise the up-regulation or down-regulation of gene expression in stem cells in response to mechanical manipulation of the stem cells as described herein. Through the manipulation of gene expression patterns or levels, the stem cells may be induced to follow particular differentiation pathways such as described herein.
Preferentially, the method of the invention comprises the remote manipulation of cells and/or of agonising or autagonising ion channels, e.g. manipulation from outside the body., i.e. remote mechanical activation. The method ofthe invention may be utilised in relation to a variety of cells which are known per se. However, preferentially, the method is suitable for use with mammalian stem cells.
The method of the invention may be utilised in connection with any conventionally known ion channels within the cell which are hereinbefore described. The method is especially suited for use in mechanosensitive ion channels. Such mechanosensitive ion channels have been identified in many cell types and have been predominantly described as calcium or potassium ion channels, although it should be understood that the method of the invention is not limited to use in relation to calcium or potassium ion channels. By way of example only, one such channel which has been well characterised at the molecular level and at the functional level in neuronal cells is the chromosomal gene TREK-1, which is part of the 2P K+ channel family. TREK-1 channels, have been identified in bone cells, and are known to respond to shear stress, cell swelling and membrane stretch as well as other external agents such as fatty acids and general anaesthetics.
A particular aspect of the present invention is to provide a method of manipulating mechanosensitive ion channels.
These "mechanosensitive" ion channels are present in a variety of mammalian, e.g. human, and bacterial cells and the present invention enables the cells to be selectively activated in the body and/or in cell cultures, see, for example, Sokabe, M, F Sachs, A Jing (1991) Quantitative video microscopy of patch clamped membranes: Stress, strain, capacitance, and stretch channel activation. Biophys J. 59: 722-728; Stewart, Z, B Martinac and J Dobson (2000) Evidence for mechanosensitive transmembrane ion channels of small conductance in magnetotactic bacteria. Electro- and Magnetobiol. 19: 81-89. As these channels are instrumental in normal cellular function and play a particularly important role in, for example, the production of bone and connective tissue or activation ofthe peripheral nervous system, the ability to manipulate them remotely, e.g. from outside the body, is especially advantageous an provides applications in, ter alia, pain relief, e.g. anaesthetics, therapeutics, tissue engineering and repair and cancer therapy.
In a further aspect of the invention the method may also be suitable for use with conventionally non mechanosensitive cells and/or ion channels by the transfection of channels into cells which may otherwise be otherwise non-responsive.
All ion channels open and close (i.e. change conformational state) in response to forces and this is the principle behind ion channel activation. In the case of mechanosensitive ion channels, the force results in membrane deformation, triggering the opening of the channel. Noltage-gated and ligand-gated ion channels are also "mechanoresponsive" in that they respond to mechanical stresses on the ion channel generated by coulomb forces (in the case of voltage-gated channels) and binding forces (in the case of ligand-gated channels). As such, all ion channels can be activated by the method described herein provided that the magnetisable particle is coupled, either directly or indirectly, to the mechanoresponsive region ofthe channel protein.
Thus, in one aspect of the present invention the ion channel is a voltage-gated ion channel, alternatively, the ion channel is a ligand-gated ion channel.
A wide variety of particles may be used in the method of the invention. The magnetisable particle used in the method ofthe invention may be inherently magnetic or, alternatively, may be one which reacts in a magnetic field. Generally, any magnetic material may be used, however, by the term magnetic we mean, for example, a material which is paramagnetic superparamagnetic, ferromagnetic and/or antiferromagnetic, examples of which include elemental iron (Fe), or an compound, e.g. an iron salt, such as, magnetite (Fe3O4), maghemite (γFe2O3), and greigite (Fe3S4), or a chromium compound, e.g. a chromium salt, such as chromium oxide (CrO2), or any combination thereof. Preferably the magnetic material comprises particles, e.g. nanoparticles, which comprises a magnetic core with a biocompatible coating. Thus, such preferred particles are nanoparticles and especially nanoparticles having a core and, e.g. a silica shell enveloping the core. However, also porous particles with multiple magnetic centres within the pores. An example of such particles are those nanoparticles described in US Patent No. 6,548,264 which is incorporated herein by reference. Thus, the prior art nanoparticles may have a mean size of less than 1 micron, each of said nanoparticles comprising (a) a core comprising a magnetisable particle and (b) a silica shell enveloping the core, wherein the magnetisable particle is a magnetic material as hereinbefore described.
The micro- and nano- particles (intended to be attached to the cells) will generally be substantially spherical or elliptical. The size of the particles may vary according, inter alia, to the nature of the magnetisable material, the application, etc. However, an example of particles may be nanoparticles can having a mean size, e.g. diameter, of 5000 nm or less, e.g. from 1 nm to 5000 nm, preferably from 1 nm to 1000 nm, more preferably from 1 nm to 300 nm, or from 2 nm to 10 nm).
The particles for attachment to the cells may be coated or uncoated and single or multi-domain. Examples of suitable particles include, but are not limited to:
(i) Coated magnetic microspheres (d = 4 μm) available from Spherotech, Inc. These microspheres consist of a magnetically blocked core - coated by a polymer.
(ii) Single-domain, ferrite-doped silica nanoparticles with tunable size (d = 50- 300 nm) and narrow size distribution.
In the method of the invention the ion channels may be activated by attaching the magnetisable particles as hereinbefore described to specific regions of the cellular membrane and/or to specific "receptors" on the ion channels themselves. Thus, the mechanical forces required to activate the channels can then be applied remotely by a magnetic field acting on these magnetic particles. In particular the method of the invention comprises modifying a magnetisable particle as hereinbefore described by tagging the particle with one or more specific antibodies or protein binding motifs which recognise key cellular elements within a cell. These include transmembrane adhesion molecules, such as integrins, cadherins, selectins, and immunoglobulins or dispersed membrane adhesion proteins such as RGD (argiiiine-glycine-aspartate), see, for example, . J. Chen, B. Fabry, E. L. Schiffrin, and N. Wang (2001) Twisting integrin receptors increases endothelin-1 gene expression in endothelial cells Am JPhysiol Cell Physiol. 280: 1475-84 ; A. R. Bausch, U. Hellerer, M. Essler, M. Aepfelbacher, and E. Sackmann (2001) Rapid stiffening of integrin receptor-actin linkages in endothelial cells stimulated with thrombin: a magnetic bead microrheology study Biophys J 80: 2649-57 ; Cartmell. SH, J Dobson, S Nerschueren, A El Haj (2002) Development of magnetic particle techniques for long-term culture of bone cells with intermittent mechanical activation.7EEE Transactions on NanoBioscience 1: 92-97.
The method ofthe invention is especially advantageous because it provides a method of treatment of a variety of disorders. Indeed the invention provides a method of treatment which is applicable to any disorder in which one or more ion channels play a role. In addition, the invention provides a method for potential control of ion channel activation including pain relief, e.g. an anaesthetic role.
Thus according to the invention we provide a method of treatment of a patient suffering from a disorder in which an ion channel plays a role which comprises the administration to such a patient of magnetisable nanoparticles as hereinbefore described and manipulating those particles using a magnetic field.
The method of treatment as hereinbefore described should not be considered to be limited, but it is especially advantageous in tissue and/or bone repair. The method of treatment can be to facilitate further treatment by providing a method of pain relief, e.g. for localised anaesthesia, to targeted regions ofthe body. The nature of such cells may vary depending upon the nature ofthe tissue of interest. For example, the cells may be ligamentum cells for growing new ligaments, tenocytes for growing new tendon. Alternatively, the cells may be chondrocytes and/or other stromal cells, such as chondrocyte progenitor cells.
Thus the method of the invention may include the regeneration of tissue or the generation of artificial tissue, such as skin, cartilage, ligament, tendon, muscle or bone.
Alternatively the method may comprise wound healing and/or tissue adhesion.
In a preferred embodiment the method may comprise bone repair and/or bone growth.
In a yet further alternative the method of the invention may include, for example, dental applications and/or veterinary applications.
The method also may be used as a mechanism for selectively killing cells (such as tumour cells) in vivo. In this case, magnetisable particles are attached to the target cell membrane or ion channel protein and a magnetic field is applied to the in vivo target region. The rapid, cyclic opening and closing (via the application of a time varying magnetic field), and/or the holding open (via the application of a static magnetic field) of ion channels in the cell membrane allows ions (such as Ca""") to flood the cell, inducing osmotic shock and, consequently, cell death.
Thus, according to this aspect of the invention we also provide a method of destroying cells or inhibiting cell growth which comprises agonising or antagonising ion channels within a cell which by the association of a magnetisable particle with a cell. The method may comprise a method of inducing osmotic shock to a cell, e.g. by agonising or antagonising ion channels within a cell by the association of a magnetisable particle with a cell. The method is especially useful in the treatment or alleviation of a tumour cell, e.g. a cancer cell.
Thus, the method may comprise the killing of cells by holding ion channels open with a targeted static magnetic field. Alternatively, the method may comprise the killing of cells via cyclically opening and closing ion channels with a targeted, time- varying magnetic field.
In the methods of the invention the magnetic field may be varied depending upon, inter alia, the nature of the disorder to be treated, but may be, for example, at a frequency of from 0.1 to 10 Hz. But, frequencies outside this range can also be used. The magnetic field will typically have a flux density in the order of (but not limited to) 10 mT to 1400 mT.
In the method of the invention the magnetic field may be generated outside the body for the case of in vivo applications, and may be provided by a permanent magnet or an electromagnet. The magnetic field may be a constant or a variable field, e.g. a permanent magnet may be moved relative to the cells. In the case of an electromagnet, a magnetic field may be generated by provision of appropriate electric current levels to the electromagnetic, optionally, in combination with alternating current.
According to a yet further aspect of the invention we provide a method of inducing a therapeutic effect in a cell which comprises agonising or antagonising ion channels within the cell by the association of a magnetisable particle with the cell and magnetically manipulating the magnetisable particle.
In addition we provide a method of treatment which comprises the administration of a therapeutically active agent which may be administered simultaneously, separately or sequentially with a magnetisable particle whilst agonising or antagonising ion channels vvithin the cell.
We also provide a method of targeting a therapeutically active agent to a cell which comprises agonising or antagonising ion channels within the cell by the association of a magnetisable particle with the cell, magnetically manipulating the magnetisable particle and simultaneously, separately or sequentially administering the therapeutically active agent.
According to a yet further aspect of the invention we also provide the use of a magnetisable particle in a method of magnetically manipulating cells in vivo
The use may comprise ex vivo manipulation of an in vivo process. More particularly, the invention provides the use of a magnetisable particle in the manufacture of a system for magnetically manipulating a cell which system comprises the association of a magnetisable particle with a cell and agonising or antagonising ion channels witliin the cell.
In this aspect of the invention the magnetisable particle may be associated directly with the cell. Alternatively, the use may comprise associating the magnetisable particle with an antibody, enzyme, etc., which is subsequently associated with the cell.
When the use of the invention comprises intracellular association. By way of example only, for TREK-1, the particle(s) may be associated with the N-terminus region of the ion channel. Alternatively, the particle(s) may be associated with the COOH terminus region ofthe ion channel.
The use ofthe invention may comprise the manipulation of mammalian cells or other cell types, such as bacterial cells, plant cells, etc. The use may be an in vitro use or an in vivo use, although an in vivo use is preferred. Preferentially, the use of the invention comprises the remote manipulation of cells and/or of agonising or autagonising ion channels, e.g. manipulation from outside the body, i.e. remote mechanical activation.
The use of the invention may be utilised in relation to a variety of cells, which are known per se. However, preferentially, the use is suitable for use with mammalian somatic cells, for example, bone, cartilage, muscle (skeletal and cardiac) lymphatic cells, endocrine cells, urinary system cells, cells relating to the reproduction system, neuronal cells and tumour cells.
The use of the invention may be utilised in connection with any conventionally known ion channels within the cell, which is hereinbefore described. The use is especially suited for use in mechanosensitive ion channels hereinbefore described.
A particular aspect of the present invention is to provide the use in the manufacture of a system for manipulating mechanosensitive ion channels.
In a further aspect of the invention the use may also be suitable for use with conventionally non mechanosensitive cells and/or ion channels by the transfection of channels into cells which may otherwise be otherwise non-responsive.
In one aspect ofthe present invention the ion channel is a voltage-gated ion channel, alternatively, the ion channel is a ligand-gated ion channel.
A wide variety of particles may be used in the use of the invention. Generally, any magnetisable material may be used, examples of which include elemental iron (Fe), or an iron compound, e.g. an iron salt, such as, magnetite (Fe3O4), maghemite (γFe2O3), and greigite (Fe3S4), or a chromium compound, e.g. a chromium salt, such as, chromium oxide (CrO2), or any combination thereof. Preferably the magnetic material comprises particles which comprises a magnetic core with a biocompatible coating. Thus, such preferred particles are nanoparticles and especially nanoparticles having a core and, e.g. a silica shell enveloping the core. However, also porous particles with multiple magnetic centres within the pores. An example of such particles are those nanoparticles described in US Patent No. 6,548,264 which is incorporated herein by reference.
In particular the use of the invention comprises modifying a magnetisable particle as hereinbefore described by tagging the particle with one or more specific antibodies or protein binding motifs which recognise key cellular elements within a cell. These include transmembrane adhesion molecules, such as integrins, cadherins, selectins, and immunoglobulins or dispersed membrane adhesion proteins such as RGD (arginine-glycine-aspartate) .
The use of the invention is especially advantageous because it provides a system suitable for use in the treatment of a variety of disorders. Indeed the invention provides the use in the manufacture of a medicament suitable for a treatment, which is applicable to any disorder in which one or more ion channels play a role. In addition, the invention provides the use for potential control of ion channel activation including pain relief, e.g. an anaesthetic role.
Thus, according to the invention we provide the use of a magnetisable particle in the manufacture of a medicament suitable for the treatment of a patient suffering from a disorder in which an ion channel plays a role which comprises the administration to such a patient of magnetisable particles as hereinbefore described and manipulating those particles using a magnetic field.
The use as hereinbefore described should not be considered to be limited, but it is especially advantageous in tissue and/or bone repair. The use can be to facilitate further treatment by providing a method of pain relief, e.g. for localised anaesthesia, to targeted regions ofthe body. The nature of such cells may vary depending upon the nature ofthe tissue of interest. For example, the cells may be ligamentum cells for growing new ligaments, tenocytes for growing new tendon. Alternatively, the cells may be chondrocytes and/or other stromal cells, such as chondrocyte progenitor cells.
Thus, the use may include the regeneration of tissue or the generation of artificial tissue, such as skin, cartilage, ligament, tendon, muscle or bone.
Alternatively the use may comprise wound healing and/or tissue adhesion.
In a preferred embodiment the use may comprise bone repair and/or bone growth.
In a yet further alternative the use ofthe invention may include, for example, dental applications and/or veterinary applications.
The use also may be used as a mechanism for selectively killing cells (such as tumour cells) in vivo as hereinbefore described.
Thus, according to this aspect of the invention we also provide the use of a magnetisable particle in the manufacture of a system for destroying cells or inhibiting cell growth which comprises agonising or antagonising ion channels within a cell which by the association of a magnetisable particle with a cell.
The use may comprise use in a method of inducing osmotic shock to a cell, e.g. by agonising or antagonising ion channels within a cell by the association of a magnetisable particle with a cell. The use in this aspect of the invention is especially useful in the treatment or alleviation of a tumour cell, e.g. a cancer cell.
Thus, the use may comprise the killing of cells by holding ion channels open with a targeted static magnetic field. Alternatively, the use may comprise the killing of cells via cyclically opening and closing ion channels with a targeted, time- varying magnetic field. According to a yet further aspect of the invention we provide the use of a magnetisable particle in the manufacture of a system for inducing a therapeutic effect in a cell which comprises agonising or antagonising ion channels within the cell by the association of a magnetisable particle with the cell and magnetically manipulating the magnetisable particle.
In addition we provide the use of a magnetisable particle in the manufacture of a system comprising a therapeutically active agent which may be administered simultaneously, separately or sequentially with the magnetisable particle whilst agonising or antagonising ion channels within the cell.
We also provide the use of a magnetisable particle in the manufacture of a system for targeting a therapeutically active agent to a cell which comprises agonising or antagonising ion channels within the cell by the association of a magnetisable particle with the cell, magnetically manipulating the magnetisable particle and simultaneously, separately or sequentially administering the therapeutically active agent.
According to a yet further aspect of the invention we provide a kit comprising a therapeutically active agent and means for associating a magnetisable particle with a cell.
It will be understood by the skilled that any conventionally known therapeutically active agent or a combination of therapeutically active agents may be utilised in the kit ofthe invention.
Thus, the kit may comprise a vessel containing a therapeutically active agent, a source of magnetisable particles and instructions for the simultaneous, sequential or separate aα'ministration thereof. The kit of the invention may also include other agents known er se. The invention may also include the use of a kit as hereinbefore described in the manufacture of a medicament. The invention will now be described by way of example only and with reference to the accompanying drawings in which Figure la) is a schematic representation ofthe structure of TREK-1 showing the three sites of 12x histidine insertions for tagging magnetic beads for mechanical manipulation;
Figure lb) illustrates primary human astrocytes with membrane bound RGD coated carboxyl ferromagnetic particles (4μm) (magnification x 1000); Figure 2 is a schematic of the TREK ion channel showing structure and location of the His. tags present in the protein. Red circles indicate the sites ofthe His tags at the three sites, the primary loop, the COOH terminus and the NH terminus;
Figure 3 is a representation of the magnetic activation of Trek-1 monitored via downstream changes in intracellular calcium; and
Figure 4 is a representation of the magnetic activation of TREK-1 induces transient rise in intracellular calcium in HEK293 T cells co-transfected with and Flashpericam.
Example 1
Targeting model system
The model system consists of a peristaltic pump connected to tubing which feeds into channels witnin agar gel blocks. The magnets can be placed at various positions in relation to the channels and the magnetic field and gradient at the target site is measured using an axial Hall probe interfaced to a gaussmeter. The magnetic fields generated by the rare earth magnets will be characterised using a Redcliffe Diagnostics MagScan field mapping system requested for this project. After each experimental run, the gel channel will be excised and assayed for cell capture using staining techniques. Magnetic particle capture will be quantified by performing Superconducting Quantum Interference Device (SQUID) magnetometry measurements on the freeze dried gel blocks. Models may be used to optimize the delivery and targeting parameters, such as magnetic field strength and geometry, magnetic particle characteristics, number of particles per cell, etc. Example 2
Non-specific membrane deformation using magnetic cytometry
Specifically, scaffolds are seeded with 106-109 BMSc dependant on scaffold size and cultured for 24 hours prior to placing within the bioreactor. Constructs are then subjected to varying magnetic loading regimes, e.g. 1 hour at 1 Hz frequency with forces ranging from 1-lOOpN per particle. These parameters are controllable and will allow optimisation of the system for varying cell types and scaffold materials. Following treatment, cells may be removed and subjected to RNA and protein analysis at varying points after activation. Using Western blotting, FACs analysis and quantitative PCR techniques assays may be conducted for osteoblastic transcription factors, such as runx 2 and osterix, alongside matrix proteins, such as osteopontin, collagen type 1, alkaline phosphatase and osteocalcin.
Example 3
Demonstration of new bone formation in animal models to validate the applicability of these magnetic micro and Nanoparticles
Animal trials of this technology support the ability to remotely activate stem cells to promote bone call differentiation and new bone formation by cells held in vivo within subcutaneous diffusion chambers using a mouse SCID model. In this way, comparisons can be made with in vitro experiments. Targeting of cells to specific tissues in vivo may also be advised.
Example 4
Demonstration of in vivo bone formation
Human-derived osteoprogenitors from mesenchymal stem cells may be used. In vivo bone formation may be assessed using the subcutaneous implant model in severely compromised immunodeficient (SCID) mice and the diffusion chamber model. This provides a rapid and robust model to validate, in vivo, the efficacy for targeting of magnetic micro- and nanoparticles and provides a clear demonstration of bone formation. The diffusion chamber assay provides unequivocal demonstration of bone formation by implanted cells as opposed to host cells. The subcutaneous implant model remains the industry standard for the assessment of skeletal tissue formation and one of us (RO) has published on the use of both the sc and DC models under a project license to RO (30/1759) for assessment of skeletal tissue engineering22. In brief, selected human osteoprogenitor cells will be implanted subcutaneously in SCID mice for four weeks while for diffusion chamber studies, cells and magnetic particle composites will be placed into each diffusion chamber and the chambers implanted intraperitoneally into athymic nude mice (MFI-nu-nu; 4-6 weeks old; Harlan UK Ltd) for 10 weeks. Thereafter, diffusion chambers will be removed, fixed overnight (95% ethanol, 4°C) and embedded undecalcified in poly(hydroxymethylmethacrylate) resin at 4°C. New bone formation will be assessed by histological techniques including frozen, paraffin and methylmethacrylate plastic sections. Assessment of cartilage and bone formed will be by histological examination using toluidine blue Giemsa, alcian blue/sirius red and Safranin-O staining. The model is currently run in Southampton under a project licence to RO (30/1759).
Example 5
Targeting of cells to specific sites in vivo
This work will focus on delivery of magnetic particle-loaded cells to specific tissue sites via intra-arterial and intravenous injection. In brief, selected and expanded mesenchymal stem cells will be loaded with magnetic particles and injected by tail vein into anesthesized MFlnu/nu mice. The cells will be localised to a specific target site using external high-gradient NdFeB magnets. Control mice also will be injected, however, no magnet will be used for targeting. Targeting efficiency will be assayed using MRI (magnetic nanoparticles are used as contrast enhancement agents in clinical MR imaging) and SQUID magnetometry analysis of dissected, freeze-dried target tissue after 4, 7 and 14 days. Example 6
Preliminary experiments using the technique described herein have been conducted on human bone-derived mesechymal stem cells [Cambrex poietics-hMSC]. Cells cultured in alpha MEM with 10% FCS and 1% antibiotics, ascorbic acid (50micrograms/ml) and beta-glycerophosphate (lOmM) (sample groups E-H) for 5 days.
Magnetic microparticles (d-4μm) were coated with a biotinylated α2/δ-l subunit of a voltage gated calcium ion channel receptor antibody. After 4 days, particles were attached to the stem cells for 40 minutes via the calcium channel receptor. After 40 minutes, the cells were exposed to a 1Hz magnetic field which applied a force of approximately 30 picoNewtons per particle (~2 particles/cell). After 2 hours 40 minutes, the particles were detached from the cells and removed by aspiration. The original culture media was returned to the samples which were then further cultured for another 24 hour period. RNA from the control and stimulated groups was collected at day five. Gene microarray analysis was performed on each of the samples. 8000 genes/sample were analysed using HG-Focus human genome chips (Affymetrix UK Ltd) in response to magnetic activation (upregulation and downregulation taken as two fold increase/decrease).
Microarray data from these experiments showed that the mechanical stimulation resulted in the downregulation of certain genes such as nerve growth factor and fibroblast growth factor (Table 1). This is an indication that the application of mechanical force using magnetic particles is guiding the stem cell differentiation away from the neuronal and fibroblast pathways. The upregulation of genes such as tetranectin in response to the mechanical force application indicates a differentiation of the cells towards an osteogenic pathway. Upregulation of genes involved in cytoskeletal reorganisation and cell adhesion proteins correlate with expected cell processes after force application.
Figure imgf000025_0001
Table 1: Results of microarray analysis showing the activity of specific genes related to differentiation and mechanical stimulation and their relative up- and downregulation compared to the non-stimulated control cultures. Symbols: upregulation and ▼ = downregulation.
P103967WO

Claims

Claims
1. A method of selectively activating and/or targeting stem cells which enables the cells to then be manipulated mechanically in a remote manner.
2. A method according to claim 1 characterised in that the remote manner is a non-contacting manner and in the case of in vivo activating/targeting specifically from outside the body.
3. A method according to claim 1 characterised in that the method comprises magnetically manipulating a stem cell in vivo or in vitro by the association of a magnetisable particle with a stem cell.
4. A method according to claim 1 characterised in that the method comprises
(i) targeting stem cells to the site of repair and/or holding the cells at that site; and
(ii) conditioning and/or differentiating in vitro and/or in vivo.
5. A method according to claim 1 characterised in that the method comprises the targeting of stem cells in vivo.
6. A method according to claim 1 characterised in that the method comprises the manipulation of human stem cells.
7. A method according to claim 1 characterised in that the method comprises tagging the stem cells with magnetisable nanoparticles which can be delivered to or held at, a particular repair site by external magnetic manipulation.
8. A method according to claim 1 characterised in that the method comprises remote activation of specific stem cell membrane receptors.
9. A method according to claim 1 characterised in that the method comprises deposition of stem cells at a site, retairring the cells at the site and remotely activating the cells in situ within a patient.
10. A method according to claim 1 characterised in that the method comprises targeting specific receptors on stem cells for remote activation of transmembrane ion channels in stem cells.
11. A method according to claim 1 characterised in that the method comprises early stage differentiation of cell types.
12. A method according to claim 1 characterised in that the method comprises targeting a variety of stem cell receptor types present in human bone marrow stem cells.
13. A method according to claim 12 characterised in that the stem cell receptor types are selected from mechano-activated ion channels e.g. K+ channels (TREK), calcium channels, integrins and surface membrane binding sites, such as RGD.
14. A method according to claim 13 characterised in that the method comprises targeting receptors for external growth factors (e.g. TGFB and BMP2) which have been shown to activate downstream transcription factors such as Runx2 and Osterix, (critical for stem cell differentiation).
15. A method according to claim 1 characterised in that the stem cells are mesenchymal stem cells.
16. A method according to claim 15 characterised in that the method comprises engraftment of human mesenchymal stem cells at the site of injury or repair.
17. A method according to claim 1 characterised in that the method provides therapeutic treatment.
18. A method according to claim 17 characterised in that the therapeutic treatment is selected from gene therapy and tissue engineering.
19. A method according to claim 18 characterised in that the site is a tissue repair site.
20. A method according to claim 1 characterised in that at the functional level the stem cell differentiated as a neuronal cell.
21. A method according to claim 1 characterised in that the method comprises stem cell binding, delivery and activation.
22. A method according to claim 1 characterised in that the method comprises using adult primary marrow human stem cells and/or human embryonic stem cells.
23. A method according to claim 1 characterised in that a bioreactor enables forces to be applied to magnetic particles attached to stem cells cultured in vitro within a multi-well 2D system or in vivo a 3D scaffold-based system.
24. A method according to claim 23 characterised in that the mesenchymal stem cells comprise populations selected from osteogenic, chondrogenic and adipogenic populations.
25. A method according to claim 1 characterised in that the method comprises magnetic activated cell sorting (MACS) with a monoclonal antibody.
26. A method according to claim 25 characterised in that the monoclonal antibody is STRO-1.
27. A method according to claim 23 characterised in that the method includes BMSc culture in monolayer, using 3D scaffolds composed of biodegradable polymers.
28. A method according to claim 27 characterised in that the biodegradable polymer is selected from polylactic acid (PLLA) and a collagen gel.
29. A method according to claim 1 characterised in that the method comprises ex vivo manipulation of an in vivo process.
30. A method according to claim 1 characterised in that the method comprises the activation and/or targeting of a magnetisable particle with a stem cell.
31. A method of magnetically manipulating a stem cell which comprises the association of a magnetisable particle with a cell characterised in that the method comprises agonising or antagonising ion channels within a cell by the association of a magnetisable particle with a cell.
32. A method according to claims 1 or 31 characterised in that the method includes a differentiation step.
33. A method according to claims 1 or 31 characterised in that the magnetisable particle is associated directly with the stem cell.
34. A method according to claims 1 or 31 characterised in that the method comprises associating the magnetisable particle with an antibody or an enzyme which antibody or enzyme is subsequently associated with the stem cell.
35. A method according to claims 1 or 31 characterised in that the method comprises the introduction of a particle into a stem cell or the attachment of a particle to a stem cell.
36. A method according to claim 35 characterised in that particles are associated intracellularly or extracellularly.
37. A method according to claim 36 characterised in that particles are associated intracellularly.
38. A method according to claim 37 characterised in that the intracellular association comprises association with an internal binding site.
39. A method according to claims 1 or 31 characterised in that the method comprises manipulating a mechanosensitive ion channel in a stem cell characterised in that the method comprises the association of a magnetisable particle with an ion channel, either directly or indirectly.
40. A method according to claim 39 characterised in that particles are associated with the N-teiminal region ofthe ion channel.
41. A method according to claim 39 characterised in that particles are associated with the COOH terminal region ofthe ion channel.
42. A method according to claim 39 characterised in that the method comprises the remote manipulation of stem cells and/or of agonising or autagonising an ion channel remotely.
43. A method according to claims 1 or 31 characterised in that the method comprises the utilisation of stem cells known to respond to shear stress, cell swelling and membrane stretch and/or external agents.
44. A method according to claim 43 characterised in that the external agent is a fatty acid or a general anaesthetic.
45. A method according to claims 1 or 31 characterised in that the method is incorporated in an application of pain relief, anaesthesia, therapeutics, tissue engineering and repair and/or cancer therapy.
46. A method according to claim 45 characterised in that the stem cell is differentiated to connective or neuronal tissue.
47. A method according to claim 45 characterised in that the stem cell is differentiated to bone, neurons, cardiac cells or any combination thereof.
48. A method according to claim 39 characterised in that the ion channel is a mechanosensitive ion channel.
49. A method according to claim 39 characterised in that the mechanosensitive ion channel has been transfected into a cell. '
50. A method according to claim 39 characterised in that the method comprises the use of force resulting in membrane deformation, triggering the opening of the channel or Noltage-gated and ligand-gated ion channels.
51. A method according to claim 50 characterised in that the ion channel is a voltage-gated ion channel.
52. A method according to claim 50 characterised in that the ion channel is a ligand-gated ion channel.
53. A method according to claim 39 characterised in that the ion channel is selected from the group a including sodium channel, potassium channel, calcium channel, chloride channel and a non-selective cation channel or any combination thereof.
54. A method according to claim 53 characterised in that the ion channel is selected from a calcium or a potassium ion channel.
55. A method according to claim 54 characterised in that the ion channel is a potassium ion channel.
56. A method according to claim 55 characterised in that the potassium channel is a TREK-1 channel.
57. A method according to claim 56 characterised in that the method comprises the utilisation of TREK-1 channels in bone cells.
58. A method according to claims 1 or 31 characterised in that the method comprises targeting using an external high gradient rare earth magnet.
59. A method according to claim 58 characterised in that the rare earth magnet is a NdFeB magnet.
60. A method according to claims 1 or 31 characterised in that the magnets produce high field/gradient products which exert a translational force on the magnetic particles loaded onto the cells, holding them at the target site according to the equation: Fmag = (X2- Xl)N- B(VB) o
61. A method according to claims 1 or 31 characterised in that the activation comprises remote mechanical activation achieved using a magnetic conditioning bioreactor.
62. A method according to claims 1 or 31 characterised in that the magnetisable particle used in the method of the invention may be inherently magnetic or, alternatively, may be one which reacts in a magnetic field.
63. A method according to claims 1 or 31 characterised in that the magnetisable particle is magnetic.
64. A method according to claim 63 characterised in that the magnetic material is paramagnetic superparamagnetic, ferromagnetic and/or antiferromagnetic,
65. A method according to claim 62 characterised in that the magnetisable material is selected from the group which includes elemental iron (Fe), or a compound thereof, and a chromium compound, or a combination thereof.
66. A method according to claim 65 characterised in that the iron compound is an iron salt.
67. A method according to claim 66 characterised in that the iron salt is selected from the group which includes magnetite (Fe3O4), maghemite (γFe2O3) and greigite
(Fe3S4), or any combination thereof.
68. A method according to claim 65 characterised in that the chromium compound is a chromium salt.
69. A method according to claim 68 characterised in that the chromium salt is chromium oxide (CrO2).
70. A method according to claim 63 characterised in that the magnetic material comprises particles which comprises a magnetic core with a biocompatible coating.
71. A method according to claim 70 characterised in that the biocompatible magnetic nanoparticles comprise a magnetite (Fe O4) and/or maghemite (Fe2O3) core with either a silica, dextran, or PNA coating.
72. A method according to claims 1 or 31 characterised in that the particle is a nanoparticle.
73. A method according to claim 72 characterised in that the nanoparticles have a particle size of from lnm to 1 Oμm.
74. A method according to claim 73 characterised in that the particles have a mean size of 5000 nm or less.
75. A method according to claim 74 characterised in that the particles have a mean size of from 1 nm to 5000 nm.
76. A method according to claim 72 characterised in that the magnetic nanoparticles have a particle size of from lOnm up to a few microns.
77. A method according to claims 1 or 31 characterised in that the coating is functionalized and crosslinked to membrane attachment motifs.
78. A method according to claims 1 or 31 characterised in that the magnetic nanoparticles are modified so as to customise particle intemalization frequency, binding efficiency, stability and binding on cell viability and function.
79. A method according to claim 78 characterised in that the modification includes customisation of internal binding sites as well as sites on the outer membrane.
80. A method according to claim 71 characterised in that the particle has a core and a silica shell enveloping the core.
81. A method according to claim 80 characterised in that the particle is selected from those comprising (a) a core comprising a magnetisable particle and (b) a silica shell enveloping the core.
82. A method according to claim 70 characterised in that the particle is a porous particle with multiple magnetic centre within the pores.
83. A method according to claims 1 or 31 characterised in that the method comprises the application of a remote magnetic field on the magnetisable particles.
84. A method according to claim 34 characterised in that the particle is tagged with one or more specific antibodies or protein binding motifs which recognise key cellular elements within a cell.
85. A method according to claim 84 characterised in that the specific antibodies or protein binding motifs are selected from transmembrane extracellular matrix molecules, adhesion molecules or dispersed membrane adhesion proteins or extracellular matrix proteins.
86. A method according to claim 85 characterised in that the transmembrane adhesion molecules are selected from integrins, cadherins, selectins, and immunoglobulins.
87. A method according to claim 86 characterised in that the specific antibodies or protein binding motifs are selected from dispersed membrane adhesion proteins.
88. A method according to claim 87 characterised in that the dispersed membrane adhesion protein is RGD (arginine-glycine-aspartate).
89. A method of treatment of a patient suffering from a disorder in which an ion channel plays a role which comprises the administration to such a patient of magnetisable particles and manipulating stem cell ion channels or the stem cells using a magnetic field external to the body.
90. A method of treatment or alleviation of a tumour cell which comprises a method according to claim 89.
91. A method according to claim 90 characterised in that the tumour cell is a cancer cell.
92. A method of treatment of a patient according to claim 91 characterised in that the method comprises the killing of cells via holding ion channels open with a targeted static magnetic field.
93. A method of treatment of a patient according to claim 91 characterised in that the method comprises the killing of cells via cyclically opening and closing ion channels with a targeted, time-varying magnetic field.
94. A method of treatment according to claim 91 in which a disorder may involve a number of tissues in the body where ion channels play a key role in normal cellular homeostasis.
95. A method according to claim 94 characterised in the cells are cardiac muscle cells.
96. A method according to claim 94 characterised in that the method comprises the treatment of hypertension.
97. A method according to claim 94 characterised in that the method comprises pain relief.
98. A method according to claim 97 characterised in that the method comprises anaesthesia.
99. A method according to claim 98 characterised in that the anaesthesia is localised.
100. A method of treatment of a patient according to claim 89 characterised in that the method comprises tissue and/or bone repair.
101. A method of treatment according to claim 100 characterised in that the cells are selected from ligamentum cells, tenocytes, chondrocytes and other stromal cells (such as chondrocyte progenitor cells).
102. A method of treatment according to claim 100 characterised in that the method comprises the regeneration of tissue or the generation of artificial tissue, such ! as skin, cartilage, ligament, tendon, muscle or bone.
103. A method of treatment according to claim 100 characterised in that the method comprises the remote activation of ion channels.
104. A method of treatment according to claim 100 characterised in that the method comprises wound healing and/or tissue adhesion.
105. A method of treatment according to claim 100 characterised in that the method comprises bone repair and/or bone growth.
106. A method of treatment according to claim 89 characterised in that the method comprises a dental or veterinary application.
107. A method of treatment according to claim 98 characterised in that the method establishes localised anaesthesia through the action of ion channel modulation by a magnetic field external to the body.
108. A method of treatment according to claim 89 characterised in that the method comprises the use of a magnetic field at a frequency of from 0.1 to 10 Hz.
109. A method of treatment according to claim 89 characterised in that the method comprises the use of a magnetic field will typically have a flux density of from 10 mT to 1400 mT.
110. A method of inducing a therapeutic effect in a stem cell which comprises agonising or antagonising ion channels within the cell by the association of a magnetisable particle with the cell and magnetically manipulating the magnetisable particle.
111. A method of treatment which comprises the administration of a therapeutically active agent which may be administered simultaneously, separately or sequentially with a magnetisable particle whilst agonising or antagonising ion channels within a stem cell.
112. A method of targeting a therapeutically active agent to a stem cell which comprises agonising or antagonising ion channels within the cell by the association of a magnetisable particle with the cell, magnetically manipulating the magnetisable particle and simultaneously, separately or sequentially administering the therapeutically active agent.
113. The use of a magnetisable particle in a method of magnetically manipulating a stem cell wherein the method comprises the association of a magnetisable particle with a cell.
114. The use according to claim 113 characterised in that the use comprises selectively activating and/or targeting stem cells which enables the cells to then be manipulated mechanically in a remote manner.
115. The use according to claim 113 characterised in that the remote manner is a non-contacting manner and in the case of in vivo activating/targeting specifically from outside the body.
116. The use according to claim 113 characterised in that the use comprises magnetically manipulating a stem cell in vivo or in vitro by the association of a magnetisable particle with a stem cell.
117. The use according to claim 113 characterised in that the use comprises
(i) targeting stem cells to the site of repair and/or holding the cells at that site; and
(ii) conditioning and/or differentiating in vitro and/or in vivo.
118. The use according to claim 113 characterised in that the use comprises the targeting of stem cells in vivo.
119. The use according to claim 113 characterised in that the use comprises the manipulation of human stem cells.
120. The use according to claim 113 characterised in that the use comprises tagging the stem cells with magnetisable nanoparticles which can be delivered to or held at, a particular repair site by external magnetic manipulation.
121. The use according to claim 113 characterised in that the use comprises remote activation of specific stem cell membrane receptors.
122. The use according to claim 113 characterised in that the use comprises deposition of stem cells at a site, retaining the cells at the site and remotely activating the cells in situ within a patient.
123. The use according to claim 113 characterised in that the use comprises targeting specific receptors on stem cells for remote activation of transmembrane ion channels in stem cells.
124. The use according to claim 113 characterised in that the use comprises early stage differentiation of cell types.
125. The use according to claim 113 characterised in that the use comprises targeting a variety of stem cell receptor types present in human bone marrow stem cells.
126. The use according to claim 125 characterised in that the stem cell receptor types are selected from mechano-activated ion channels e.g. K+ channels (TREK), calcium channels, integrins and surface membrane binding sites, such as RGD.
127. The use according to claim 126 characterised in that the use comprises targeting receptors for external growth factors (e.g. TGFB and BMP2) which have been shown to activate downstream transcription factors such as Runx2 and Osterix, (critical for stem cell differentiation).
128. The use according to claim 1 characterised in that the stem cells are mesenchymal stem cells.
129. The use according to claim 128 characterised in that the use comprises engraftment of human mesenchymal stem cells at the site of injury or repair.
130. The use according to claim 113 characterised in that the use provides therapeutic treatment.
131. The use according to claim 130 characterised in that the therapeutic treatment is selected from gene therapy and tissue engineering.
132. The use according to claim 131 characterised in that the site is a tissue repair site.
133. The use according to claim 113 characterised in that the at the functional level the stem cell differentiated as a neuronal cell.
134. The use according to claim 113 characterised in that the use comprises stem cell binding, delivery and activation.
135. The use according to claim 113 characterised in that the use comprises using adult primary marrow human stem cells and/or human embryonic stem cells.
136. The use according to claim 113 characterised in that the bioreactor enables forces to be applied to magnetic particles attached to stem cells cultured in vitro within a multi-well 2D system or in vivo a 3D scaffold-based system.
137. The use according to claim 136 characterised in that the mesenchymal stem cells comprise populations selected from osteogenic, chondrogenic and adipogenic populations.
138. The use according to claim 1 characterised in that the use comprises magnetic activated cell sorting (MACS) with a monoclonal antibody.
139. The use according to claim 138 characterised in that the monoclonal antibody is STRO-1.
140. The use according to claim 136 characterised in that the use includes BMSc culture in monolayer, using 3D scaffolds composed of biodegradable polymers.
141. The use according to claim 140 characterised in that the biodegradable polymer is selected from polylactic acid (PLLA) and a collagen gel.
142. The use according to claim 113 characterised in that the use comprises ex vivo manipulation of an in vivo process.
143. The use according to claim 113 characterised in that the use comprises the activation and/or targeting of a magnetisable particle with a stem cell.
144. The use of a magnetisable particle in the manufacture of a therapy that comprises agonising or antagonising ion channels within a stem cell by the association ofthe magnetisable particle with a stem cell.
145. The use according to claims 113 or 144 characterised in that the use includes a differentiation step.
146. The use according to claims 113 or 144 characterised in that the magnetisable particle is associated directly with the stem cell.
147. The use according to claims 113 or 144 characterised in that the use comprises associating the magnetisable particle with an antibody or an enzyme which antibody or enzyme is subsequently associated with the stem cell.
148. The use according to claims 113 or 144 characterised in that the use comprises the introduction of a particle into a stem cell or the attachment of a particle to a stem cell.
149. The use according to claim 148 characterised in that particles are associated intracellularly or extracellularly.
150. The use according to claim 149 characterised in that particles are associated intracellularly.
151. The use according to claim 150 characterised in that the intracellular association comprises association with an internal binding site.
152. The use according to claim 113 or 144 characterised in that the use comprises manipulating a mechanosensitive ion channel in a stem cell characterised in that the use comprises the association of a magnetisable particle with an ion channel, either directly or indirectly.
153. The use according to claim 152 characterised in that particles are associated with the N-terminal region of the ion channel.
154. The use according to claim 152 characterised in that particles are associated with the COOH terminal region ofthe ion channel.
155. The use according to claim 152 characterised in that the use comprises the remote manipulation of stem cells and/or of agonising or autagonising an ion channel remotely.
156. The use according to claim 113 or 144 characterised in that the use comprises the utilisation of stem cells known to respond to shear stress, cell swelling and membrane stretch and/or external agents.
157. The use according to claim 156 characterised in that the external agent is a fatty acid or a general anaesthetic.
158. The use according to claim 113 or 144 characterised in that the use is incorporated in an application of pain relief, anaesthesia, therapeutics, tissue engineering and repair and/or cancer therapy.
159. The use according to claim 158 characterised in that the stem cell is differentiated to connective or neuronal tissue.
160. The use according to claim 158 characterised in that the stem cell is differentiated to bone, neurons, cardiac cells or any combination thereof.
161. The use according to claim 152 characterised in that the ion channel is a mechanosensitive ion channel.
162. The use according to claim 152 characterised in that the mechanosensitive ion channel has been transfected into a cell.
163. The use according to claim 152 characterised in that the use comprises the use of force resulting in membrane deformation, triggering the opening of the channel or Noltage-gated and ligand-gated ion channels.
164. The use according to claims 163 characterised in that the ion channel is a voltage-gated ion channel.
165. The use according to claims 163 characterised in that the ion channel is a ligand-gated ion channel.
166. The use according to claim 152 characterised in that the ion channel is selected from the group a including sodium channel, potassium channel, calcium channel, chloride channel and a non-selective cation channel or any combination thereof.
167. The use according to claim 166 characterised in that the ion channel is selected from a calcium or a potassium ion channel.
168. The use according to claim 167 characterised in that the ion channel is a potassium ion channel.
169. The use according to claim 168 characterised in that the potassium channel is a TREK-1 channel.
170. The use according to claim 169 characterised in that the use comprises the utilisation of TREK-1 channels in bone cells.
171. The use according to claims 113 or 144 characterised in that the use comprises targeting using an external high gradient rare earth magnet.
172. The use according to claim 171 characterised in that the rare earth magnet is a NdFeB magnet.
173. The use according to claims 113 or 144 characterised in that the magnets produce high field/gradient products which exert a translational force on the magnetic particles loaded onto the cells, holding them at the target site according to the equation:
Figure imgf000046_0001
174. The use according to claims 113 or 144 characterised in that the activation comprises remote mechanical activation achieved using a magnetic conditioning bioreactor.
175. The use according to claims 113 or 144 characterised in that the magnetisable particle used in the use ofthe invention may be inherently magnetic or, alternatively, may be one which reacts in a magnetic field.
176. The use according to claims 113 or 144 characterised in that the magnetisable particle is magnetic.
177. The use according to claim 176 characterised in that the magnetic material is paramagnetic superparamagnetic, ferromagnetic and/or antiferromagnetic,
178. The use according to claim 175 characterised in that the magnetisable material is selected from the group which includes elemental iron (Fe), or a compound thereof, and a chromium compound, or a combination thereof.
179. The use according to claim 178 characterised in that the iron compound is an iron salt.
180. The use according to claim 179 characterised in that the iron salt is selected from the group which includes magnetite (Fe3O4), maghemite (γFe2O ) and greigite (Fe3S4), or any combination thereof.
181. The use according to claim 178 characterised in that the chromium compound is a chromium salt.
182. The use according to claim 181 characterised in that the chromium salt is chromium oxide (CrO2).
183. The use according to claim 176 characterised in that the magnetic material comprises particles which comprises a magnetic core with a biocompatible coating.
184. The use according to claim 183 characterised in that the biocompatible magnetic nanoparticles comprise a magnetite (Fe3O4) and/or maghemite (Fe2O3) core with either a silica, dextran, or PNA coating.
185. The use according to claims 113 or 144 characterised in that the particle is a nanoparticle.
186. The use according to claim 185 characterised in that the nanoparticles have a particle size of from lnm to lOμm.
187. The use according to claim 187 characterised in that the particles have a mean size of 5000 nm or less.
188. The use according to claim 187 characterised in that the particles have a mean size of from 1 nm to 5000 nm.
189. The use according to claim 185 characterised in that the magnetic nanoparticles have a particle size of from lOnm up to a few microns.
190. The use according to claims 113 or 144 characterised in that the coating is functionalized and crosslinked to membrane attachment motifs.
191. The use according to claims 113 or 144 characterised in that the magnetic nanoparticles are modified so as to customise particle intemalization frequency, binding efficiency, stability and binding on cell viability and function.
192. The use according to claim 191 characterised in that the modification includes customisation of internal binding sites as well as sites on the outer membrane.
193. The use according to claim 184 characterised in that the particle has a core and a silica shell enveloping the core.
194. The use according to claim 193 characterised in that the particle is selected from those comprising (a) a core comprising a magnetisable particle and (b) a silica shell enveloping the core.
195. The use according to claim 183 characterised in that the particle is a porous particle with multiple magnetic centre within the pores.
196. The use according to claims 113 or 144 characterised in that the use comprises the application of a remote magnetic field on the magnetisable particles.
197. The use according to claim 147 characterised in that the particle is tagged with one or more specific antibodies or protein binding motifs which recognise key cellular elements within a cell.
198. The use according to claim 197 characterised in that the specific antibodies or protein binding motifs are selected from transmembrane extracellular matrix molecules, adhesion molecules or dispersed membrane adhesion proteins or extracellular matrix proteins.
199. The use according to claim 198 characterised in that the transmembrane adhesion molecules are selected from integrins, cadherins, selectins, and immunoglobulins .
200. The use according to claim 199 characterised in that the specific antibodies or protein binding motifs are selected from dispersed membrane adhesion proteins.
201. The use according to claim 200 characterised in that the dispersed membrane adhesion protein is RGD (arginine-glycine-aspartate).
202. The use of a magnetisable particle in association with a stem cell in the manufacture of a therapy for the treatment of a patient suffering from a disorder in which an ion channel plays a role which comprises the administration to such a patient of magnetisable particles and manipulating the stem cell ion channels or the stem cells using a magnetic field external to the body.
203. The use of a magnetisable particle in the manufacture of a system for targeting a therapeutically active agent to a cell which comprises agonising or antagonising ion channels within the cell by the association of a magnetisable particle with the cell, magnetically manipulating the magnetisable particle and simultaneously, separately or sequentially administering the therapeutically active agent.
204. A kit comprising a therapeutically active agent and means for associating a magnetisable particle with a cell.
205. A method or use substantially as described with reference to the accompanying drawings.
PCT/GB2004/005156 2003-12-18 2004-12-08 Stem cell targeting using magnetic particles WO2005059118A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
AU2004299704A AU2004299704A1 (en) 2003-12-18 2004-12-08 Method
US10/596,594 US8469034B2 (en) 2003-12-18 2004-12-08 Stem cell targeting and activation using magnetic particles
CA002550084A CA2550084A1 (en) 2003-12-18 2004-12-08 Stem cell targeting using magnetic particles
EP04805976A EP1699917A2 (en) 2003-12-18 2004-12-08 Stem cell targeting using magnetic particles
JP2006544539A JP2007518710A (en) 2003-12-18 2004-12-08 Method

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB0329310.7A GB0329310D0 (en) 2003-12-18 2003-12-18 Method
GB0329310.7 2003-12-18

Publications (2)

Publication Number Publication Date
WO2005059118A2 true WO2005059118A2 (en) 2005-06-30
WO2005059118A3 WO2005059118A3 (en) 2006-05-26

Family

ID=30471287

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2004/005156 WO2005059118A2 (en) 2003-12-18 2004-12-08 Stem cell targeting using magnetic particles

Country Status (9)

Country Link
US (1) US8469034B2 (en)
EP (1) EP1699917A2 (en)
JP (1) JP2007518710A (en)
KR (1) KR20060124654A (en)
CN (1) CN1898378A (en)
AU (1) AU2004299704A1 (en)
CA (1) CA2550084A1 (en)
GB (1) GB0329310D0 (en)
WO (1) WO2005059118A2 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005123907A2 (en) * 2004-06-15 2005-12-29 Baxter International Inc. Ex-vivo application of solid microparticulate therapeutic agents
WO2007113572A1 (en) * 2006-04-03 2007-10-11 Keele University Targeted therapy
JP2007269770A (en) * 2006-03-09 2007-10-18 Mitsubishi Chemicals Corp Functional magnetic super-nanoparticle and use thereof
KR100792185B1 (en) 2006-06-30 2008-01-07 재단법인서울대학교산학협력재단 Method for Fixing or Mobilizing Cells Using Magnetic Nanoparticle
JP2008541734A (en) * 2005-05-30 2008-11-27 コモンウェルス サイエンティフィック アンド インダストリアル リサーチ オーガニゼイション Production and use of basement membrane particles
US7553662B2 (en) 2000-12-22 2009-06-30 Keele University Culturing tissue using magnetically generated mechanical stresses
WO2010077168A1 (en) * 2008-12-30 2010-07-08 Bryukhovetskiy Andrey Stepanovich Drug made from stem cells with reprogrammed cell signaling, method for producing said preparation and the use thereof
EP2249800A2 (en) * 2008-02-04 2010-11-17 University Of Miami Magnetic cells for localizing delivery and tissue repair
WO2011047277A3 (en) * 2009-10-15 2011-08-18 The Brigham And Women's Hospital, Inc. Release of agents from cells
CN102895652A (en) * 2011-07-27 2013-01-30 东北师范大学 Method for promoting osteoblast differentiation by using Runx2 and Osterix and application thereof
WO2013049598A1 (en) * 2011-09-28 2013-04-04 Board Of Regents Of The University Of Texas System Alternating electric current directs, enhances, and accelerates mesenchymal stem cell differentiation into osteoblasts and chondrocytes but not adipocytes
US8447379B2 (en) 2006-11-16 2013-05-21 Senior Scientific, LLC Detection, measurement, and imaging of cells such as cancer and other biologic substances using targeted nanoparticles and magnetic properties thereof
US8469034B2 (en) 2003-12-18 2013-06-25 Mica Biosystems Limited Stem cell targeting and activation using magnetic particles
US8662085B2 (en) 2010-03-02 2014-03-04 Siemens Aktiengesellschaft Magnetic nanoparticle and group of nanoparticles
US8986736B2 (en) 2003-06-24 2015-03-24 Baxter International Inc. Method for delivering particulate drugs to tissues
US8999650B2 (en) 2004-03-01 2015-04-07 Senior Scientific Llc Magnetic needle biopsy
US9044381B2 (en) 2003-06-24 2015-06-02 Baxter International Inc. Method for delivering drugs to the brain
US9095270B2 (en) 2009-11-06 2015-08-04 Senior Scientific Llc Detection, measurement, and imaging of cells such as cancer and other biologic substances using targeted nanoparticles and magnetic properties thereof
US9364443B2 (en) 2008-03-05 2016-06-14 Baxter International, Inc. Compositions and methods for drug delivery
US9964469B2 (en) 2005-02-28 2018-05-08 Imagion Biosystems, Inc. Magnetic needle separation and optical monitoring
US10194825B2 (en) 2009-11-06 2019-02-05 Imagion Biosystems Inc. Methods and apparatuses for the localization and treatment of disease such as cancer
US10952965B2 (en) 2009-05-15 2021-03-23 Baxter International Inc. Compositions and methods for drug delivery

Families Citing this family (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ITRM20030376A1 (en) 2003-07-31 2005-02-01 Univ Roma PROCEDURE FOR THE ISOLATION AND EXPANSION OF CARDIOC STAMIN CELLS FROM BIOPSIA.
GB0405552D0 (en) * 2004-03-12 2004-04-21 Magnet Attraction Ltd Methods for the targetted delivery of biological molecules
US11660317B2 (en) 2004-11-08 2023-05-30 The Johns Hopkins University Compositions comprising cardiosphere-derived cells for use in cell therapy
WO2008156904A2 (en) * 2007-04-09 2008-12-24 Children's Medical Center Corporation Systems and methods for nanomagnetic actuation of molecular cell signaling
FR2950362B1 (en) * 2009-09-21 2011-10-21 Univ Clermont Auvergne USE OF K2P POTASSIUM CHANNEL ACTIVATORS AS ANALGIC.
US9249392B2 (en) 2010-04-30 2016-02-02 Cedars-Sinai Medical Center Methods and compositions for maintaining genomic stability in cultured stem cells
US9845457B2 (en) 2010-04-30 2017-12-19 Cedars-Sinai Medical Center Maintenance of genomic stability in cultured stem cells
US9669200B2 (en) * 2010-08-06 2017-06-06 Boston Scientific Scimed, Inc. Systems and methods for the treatment of pelvic disorders including magnetic particulates
US9180172B2 (en) 2010-12-15 2015-11-10 Ams Research Corporation Treatment of Peyronies disease
US8979797B2 (en) 2010-12-16 2015-03-17 Ams Research Corporation High pressure delivery system and method for treating pelvic disorder using large molecule therapeutics
WO2013029025A1 (en) 2011-08-24 2013-02-28 The Rockefeller University Compositions and methods to modulate cell activity
WO2013059528A2 (en) * 2011-10-18 2013-04-25 City Of Hope Encapsulated diagnostics and therapeutics in nanoparticles-conjugated to tropic cells and methods for their use
US9023612B2 (en) 2012-01-13 2015-05-05 Bell Biosystems, Inc. Eukaryotic cells with artificial endosymbionts for multimodal detection
US8956873B2 (en) 2012-01-13 2015-02-17 Bell Biosystems, Inc. Host cells with artificial endosymbionts
US8828681B2 (en) 2012-01-13 2014-09-09 Bell Biosystems, Inc. Host cells with artificial endosymbionts
US10076579B2 (en) 2012-01-13 2018-09-18 Bell Biosystems, Inc. Host cells with artificial endosymbionts
WO2013184527A1 (en) 2012-06-05 2013-12-12 Capricor, Inc. Optimized methods for generation of cardiac stem cells from cardiac tissue and their use in cardiac therapy
CA2881394A1 (en) 2012-08-13 2014-02-20 Cedars-Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
WO2014145785A1 (en) 2013-03-15 2014-09-18 Bell Biosystems, Llc Host cells with artificial endosymbionts
CN103289965A (en) * 2013-07-01 2013-09-11 浙江大学 Virus AAVMTF for expressing iron nano-particles and temperature sensitive channels and use thereof
CN103333919A (en) * 2013-07-01 2013-10-02 浙江大学 Viruses AAVSTF for expressing iron nano particles and temperature-sensitive channel and application thereof
US10557120B2 (en) * 2013-07-26 2020-02-11 Kyoto Prefectural Public University Corporation Osteoblast and method for preparing same
AU2014315321A1 (en) 2013-09-03 2016-03-03 Bell Biosystems, Inc. Host cell modification with artificial endosymbionts
KR101743539B1 (en) * 2014-04-02 2017-06-08 동국대학교 산학협력단 Method For Differentiation Of Adult Stem Cells Into Nerve Cells Using High Intensity Of Electromagnetic Field
US10696728B2 (en) 2014-07-03 2020-06-30 Chunlei Liu Polypeptides, related nucleic acids, and their uses for cell modulation and treatments
CA2962052A1 (en) 2014-09-22 2016-03-31 Rensselaer Polytechnic Institute Compositions and methods to modulate cell activity
CA2962444C (en) 2014-10-03 2023-09-05 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy
ES2574655B1 (en) * 2014-11-21 2017-04-20 Universidad De Granada Elaboration of artificial tissues comprising magnetic particles
CN107847429B (en) * 2015-06-19 2021-01-22 梁平 Method for targeting or stimulating cells or organisms using nanoparticles and external field
CN106691542A (en) * 2015-07-17 2017-05-24 成昱 Stem cell mediated magnetic knife and preparation method and application of same
US11253551B2 (en) 2016-01-11 2022-02-22 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction
US11351200B2 (en) 2016-06-03 2022-06-07 Cedars-Sinai Medical Center CDC-derived exosomes for treatment of ventricular tachyarrythmias
WO2018057542A1 (en) 2016-09-20 2018-03-29 Cedars-Sinai Medical Center Cardiosphere-derived cells and their extracellular vesicles to retard or reverse aging and age-related disorders
US11759482B2 (en) 2017-04-19 2023-09-19 Cedars-Sinai Medical Center Methods and compositions for treating skeletal muscular dystrophy
US11660355B2 (en) 2017-12-20 2023-05-30 Cedars-Sinai Medical Center Engineered extracellular vesicles for enhanced tissue delivery
US11518971B2 (en) 2018-11-27 2022-12-06 Research Triangle Institute Method and apparatus for spatial control of cellular growth
CN110680831A (en) * 2019-10-30 2020-01-14 扬州大学 New application of ferroferric oxide nanoenzyme
CN114432497B (en) * 2020-10-30 2023-11-24 江千里 Micro-magnet net film and preparation method thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002032397A2 (en) * 2000-10-18 2002-04-25 Virginia Commonwealth University Intellectual Property Foundation Electroprocessing in drug delivery and cell encapsulation
WO2002051985A2 (en) * 2000-12-22 2002-07-04 Keele University Culturing tissue using magnetically generated mechanical stresses
WO2004000369A2 (en) * 2002-06-19 2003-12-31 Keele University Method of magnetically manipulating a cell with magnetisable particles

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4105017A (en) * 1976-11-17 1978-08-08 Electro-Biology, Inc. Modification of the growth repair and maintenance behavior of living tissue and cells by a specific and selective change in electrical environment
US5183336A (en) * 1992-01-21 1993-02-02 Kontes Glass Company Stirring assembly
US5486457A (en) 1993-08-25 1996-01-23 Children's Medical Center Corporation Method and system for measurement of mechanical properties of molecules and cells
US5842477A (en) * 1996-02-21 1998-12-01 Advanced Tissue Sciences, Inc. Method for repairing cartilage
US6197586B1 (en) * 1997-12-12 2001-03-06 The Regents Of The University Of California Chondrocyte-like cells useful for tissue engineering and methods
US6649408B2 (en) * 2000-03-24 2003-11-18 George Mason University Microdroplet cell culture technique
US6548264B1 (en) 2000-05-17 2003-04-15 University Of Florida Coated nanoparticles
US7553662B2 (en) * 2000-12-22 2009-06-30 Keele University Culturing tissue using magnetically generated mechanical stresses
GB0329310D0 (en) 2003-12-18 2004-01-21 Univ Keele Method

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002032397A2 (en) * 2000-10-18 2002-04-25 Virginia Commonwealth University Intellectual Property Foundation Electroprocessing in drug delivery and cell encapsulation
WO2002051985A2 (en) * 2000-12-22 2002-07-04 Keele University Culturing tissue using magnetically generated mechanical stresses
WO2004000369A2 (en) * 2002-06-19 2003-12-31 Keele University Method of magnetically manipulating a cell with magnetisable particles

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CARTMELL S H ET AL: "Development of magnetic particle techniques for long-term culture of bone cells with intermittent mechanical activation" IEEE TRANSACTIONS ON NANOBIOSCIENCE IEEE USA, vol. 1, no. 2, June 2002 (2002-06), pages 92-97, XP011077118 ISSN: 1536-1241 *
CARTMELL S H ET AL: "Development of magnetic particle techniques for long-term culture of bone cells with intermittent mechanical activation" IEEE TRANSACTIONS ON NANOBIOSCIENCE, IEEE SERVICE CENTER, PISCATAWAY, NY, US, vol. 1, no. 2, June 2002 (2002-06), pages 92-97, XP001152893 ISSN: 1536-1241 *
FROHLICH M W ET AL: "Ex Vivo Activation and Expansion of T Cells from the Peripheral Blood of Multiple Myeloma Patients Using the XcellerateTM Process" BLOOD, W.B.SAUNDERS COMPANY, ORLANDO, FL, US, vol. 100, no. 11, 16 November 2002 (2002-11-16), XP002296916 ISSN: 0006-4971 *
LEWIN M ET AL: "Tat peptide-derivatized magnetic nanoparticles allow in vivo tracking and recovery of progenitor cells" NATURE BIOTECHNOLOGY, NATURE PUBLISHING, US, vol. 18, April 2000 (2000-04), pages 410-414, XP002958421 ISSN: 1087-0156 *

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7553662B2 (en) 2000-12-22 2009-06-30 Keele University Culturing tissue using magnetically generated mechanical stresses
US9044381B2 (en) 2003-06-24 2015-06-02 Baxter International Inc. Method for delivering drugs to the brain
US8986736B2 (en) 2003-06-24 2015-03-24 Baxter International Inc. Method for delivering particulate drugs to tissues
US8469034B2 (en) 2003-12-18 2013-06-25 Mica Biosystems Limited Stem cell targeting and activation using magnetic particles
US8999650B2 (en) 2004-03-01 2015-04-07 Senior Scientific Llc Magnetic needle biopsy
WO2005123907A3 (en) * 2004-06-15 2007-08-23 Baxter Int Ex-vivo application of solid microparticulate therapeutic agents
WO2005123907A2 (en) * 2004-06-15 2005-12-29 Baxter International Inc. Ex-vivo application of solid microparticulate therapeutic agents
US8333959B2 (en) 2004-06-15 2012-12-18 Baxter International Inc. Ex-vivo application of solid microparticulate therapeutic agents
US10900872B2 (en) 2005-02-28 2021-01-26 Imagion Biosystems Inc. Magnetic needle separation and optical monitoring
US9964469B2 (en) 2005-02-28 2018-05-08 Imagion Biosystems, Inc. Magnetic needle separation and optical monitoring
JP2008541734A (en) * 2005-05-30 2008-11-27 コモンウェルス サイエンティフィック アンド インダストリアル リサーチ オーガニゼイション Production and use of basement membrane particles
JP2007269770A (en) * 2006-03-09 2007-10-18 Mitsubishi Chemicals Corp Functional magnetic super-nanoparticle and use thereof
WO2007113572A1 (en) * 2006-04-03 2007-10-11 Keele University Targeted therapy
KR100792185B1 (en) 2006-06-30 2008-01-07 재단법인서울대학교산학협력재단 Method for Fixing or Mobilizing Cells Using Magnetic Nanoparticle
US8447379B2 (en) 2006-11-16 2013-05-21 Senior Scientific, LLC Detection, measurement, and imaging of cells such as cancer and other biologic substances using targeted nanoparticles and magnetic properties thereof
EP2249800A2 (en) * 2008-02-04 2010-11-17 University Of Miami Magnetic cells for localizing delivery and tissue repair
US9078932B2 (en) 2008-02-04 2015-07-14 Emmetrope, Inc. Magnetic cells for localizing delivery and tissue repair
EP2249800A4 (en) * 2008-02-04 2014-09-24 Jeffrey L Goldberg Magnetic cells for localizing delivery and tissue repair
US9364443B2 (en) 2008-03-05 2016-06-14 Baxter International, Inc. Compositions and methods for drug delivery
US20110268774A1 (en) * 2008-12-30 2011-11-03 Andrey Stepanovich Bryukhovetskiy preparation of stem cells with reprogrammed cell signalling, a method of producing the preparation and a method of use thereof
WO2010077168A1 (en) * 2008-12-30 2010-07-08 Bryukhovetskiy Andrey Stepanovich Drug made from stem cells with reprogrammed cell signaling, method for producing said preparation and the use thereof
US10952965B2 (en) 2009-05-15 2021-03-23 Baxter International Inc. Compositions and methods for drug delivery
US8956863B2 (en) 2009-10-15 2015-02-17 The Brigham And Women's Hospital, Inc. Agents from cells
WO2011047277A3 (en) * 2009-10-15 2011-08-18 The Brigham And Women's Hospital, Inc. Release of agents from cells
US9884129B2 (en) 2009-10-15 2018-02-06 The Brigham And Women's Hospital, Inc. Release of agents from cells
US9095270B2 (en) 2009-11-06 2015-08-04 Senior Scientific Llc Detection, measurement, and imaging of cells such as cancer and other biologic substances using targeted nanoparticles and magnetic properties thereof
US10194825B2 (en) 2009-11-06 2019-02-05 Imagion Biosystems Inc. Methods and apparatuses for the localization and treatment of disease such as cancer
US8662085B2 (en) 2010-03-02 2014-03-04 Siemens Aktiengesellschaft Magnetic nanoparticle and group of nanoparticles
CN102895652A (en) * 2011-07-27 2013-01-30 东北师范大学 Method for promoting osteoblast differentiation by using Runx2 and Osterix and application thereof
US8945894B2 (en) 2011-09-28 2015-02-03 Courtney M. Creecy Alternating electric current directs, enhances, and accelerates mesenchymal stem cell differentiation into either osteoblasts or chondrocytes but not adipocytes
WO2013049598A1 (en) * 2011-09-28 2013-04-04 Board Of Regents Of The University Of Texas System Alternating electric current directs, enhances, and accelerates mesenchymal stem cell differentiation into osteoblasts and chondrocytes but not adipocytes

Also Published As

Publication number Publication date
KR20060124654A (en) 2006-12-05
CN1898378A (en) 2007-01-17
AU2004299704A1 (en) 2005-06-30
GB0329310D0 (en) 2004-01-21
US8469034B2 (en) 2013-06-25
CA2550084A1 (en) 2005-06-30
EP1699917A2 (en) 2006-09-13
JP2007518710A (en) 2007-07-12
WO2005059118A3 (en) 2006-05-26
US20110034753A1 (en) 2011-02-10

Similar Documents

Publication Publication Date Title
US8469034B2 (en) Stem cell targeting and activation using magnetic particles
Rittchen et al. Myelin repair in vivo is increased by targeting oligodendrocyte precursor cells with nanoparticles encapsulating leukaemia inhibitory factor (LIF)
Li et al. Adaptive materials based on iron oxide nanoparticles for bone regeneration
Polak et al. Nanometric agents in the service of neuroscience: manipulation of neuronal growth and activity using nanoparticles
Kopke et al. Magnetic nanoparticles: inner ear targeted molecule delivery and middle ear implant
Whatley et al. Magnetic‐directed patterning of cell spheroids
Van de Walle et al. Ever-evolving identity of magnetic nanoparticles within human cells: The interplay of endosomal confinement, degradation, storage, and neocrystallization
Ferreira Nanoparticles as tools to study and control stem cells
Funnell et al. Magnetic composite biomaterials for neural regeneration
Lopez et al. Magneto-mechanical destruction of cancer-associated fibroblasts using ultra-small iron oxide nanoparticles and low frequency rotating magnetic fields
WO2006048321A1 (en) Multimodally modified cells used as an administrable form for active substances and as diagnostic particles
Aseri et al. Magnetic nanoparticles: Magnetic nano-technology using biomedical applications and future prospects
Zou et al. MUC‐1 aptamer targeted superparamagnetic iron oxide nanoparticles for magnetic resonance imaging of pancreatic cancer in vivo and in vitro experiment
Gao et al. Magnetic field promotes migration of schwann cells with chondroitinase ABC (ChABC)-loaded superparamagnetic nanoparticles across astrocyte boundary in vitro
Xia et al. Manipulation of Schwann cell migration across the astrocyte boundary by polysialyltransferase-loaded superparamagnetic nanoparticles under magnetic field
US7625562B2 (en) Generation of cartilage using magnetizable particles
Ahn et al. Biodistribution of poly clustered superparamagnetic iron oxide nanoparticle labeled mesenchymal stem cells in aminoglycoside induced ototoxic mouse model
WO2000071169A2 (en) Magnetic resonance tracking of magnetically labeled cells
Huang et al. 3D gradient and linearly aligned magnetic microcapsules in nerve guidance conduits with remotely spatiotemporally controlled release to enhance peripheral nerve repair
Almeida et al. Magnetic triggers in biomedical applications–prospects for contact free cell sensing and guidance
Bharti et al. Nanotechnology in stem cell research and therapy
Zhang et al. Polarization of stem cells directed by magnetic field-manipulated supramolecular polymeric nanofibers
Yoo et al. In vivo fluorescence imaging of threadlike tissues (Bonghan ducts) inside lymphatic vessels with nanoparticles
Yanada et al. Effects of CD44 antibody–or RGDS peptide–immobilized magnetic beads on cell proliferation and chondrogenesis of mesenchymal stem cells
Rotherham et al. Remote magnetic actuation of cell signalling for tissue engineering

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200480038043.3

Country of ref document: CN

AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2004299704

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 547910

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 1020067012067

Country of ref document: KR

Ref document number: 2550084

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2006544539

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

ENP Entry into the national phase

Ref document number: 2004299704

Country of ref document: AU

Date of ref document: 20041208

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004299704

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2004805976

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2004805976

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020067012067

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 10596594

Country of ref document: US